151
|
Rosaian AS, Rao GN, Mohan SP, Vijayarajan M, Prabhakaran RC, Sherwood A. Regenerative Capacity of Dental Pulp Stem Cells: A Systematic Review. JOURNAL OF PHARMACY AND BIOALLIED SCIENCES 2020; 12:S27-S36. [PMID: 33149427 PMCID: PMC7595477 DOI: 10.4103/jpbs.jpbs_121_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/06/2020] [Accepted: 03/13/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES The dental pulp contains undifferentiated mesenchymal cells, blood vessels and so on, which are responsible for routine functions of a tooth. The determination of stemness and regenerative properties using biomarkers and further application in routine practice may unravel its potential. MATERIALS AND METHODS Inclusion criteria-original research articles published in English, from 2000 to 2019, were collected both manually and by electronic search from databases of Cochrane, Medline, Embase, and PubMed. Exclusion criteria-articles other than English and review manuscripts were omitted. The shortlisted articles were reviewed for specific biomarkers, to assess the regenerative potential, stemness, and lineage of dental pulp stem cells. RESULTS Of 512 articles, 64 were selected and reviewed to determine the mesenchymal, neurogenic, vasculogenic, hematopoietic, and stem cell potential. On the basis of the search analysis, a panel of markers was proposed. CONCLUSION The application of proposed markers, on a pulpectomized tissue derived from human teeth, may be helpful to determine the regenerative potential and the usefulness in regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Adlin S Rosaian
- Department of Oral and Maxillofacial Pathology and Oral Microbiology, CSI College of Dental Sciences and Research, Madurai, Tamil Nadu, India
| | - Gururaj Narayana Rao
- Department of Oral and Maxillofacial Pathology and Oral Microbiology, CSI College of Dental Sciences and Research, Madurai, Tamil Nadu, India
| | - Sunil P Mohan
- Department of Oral Pathology, Sree Anjaneya Institute of Dental Sciences, Kozhikode, Kerala, India
- Department of Stem Cells and Regenerative Medicine, Sree Anjaneya Institute of Dental Sciences, Kozhikode, Kerala, India
| | - Mahalakshmi Vijayarajan
- Department of Oral and Maxillofacial Pathology and Oral Microbiology, CSI College of Dental Sciences and Research, Madurai, Tamil Nadu, India
| | - Rebekkah C Prabhakaran
- Department of Oral and Maxillofacial Pathology and Oral Microbiology, CSI College of Dental Sciences and Research, Madurai, Tamil Nadu, India
| | - Anand Sherwood
- Department of Operative Dentistry and Endodontics, CSI College of Dental Sciences and Research, Madurai, Tamil Nadu, India
| |
Collapse
|
152
|
Cakouros D, Gronthos S. The changing epigenetic landscape of Mesenchymal Stem/Stromal Cells during aging. Bone 2020; 137:115440. [PMID: 32445894 DOI: 10.1016/j.bone.2020.115440] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/17/2020] [Accepted: 05/17/2020] [Indexed: 12/17/2022]
Abstract
There is mounting evidence in the literature that mesenchymal stromal/stem cell (MSC) like populations derived from different tissues, undergo epigenetic changes during aging, leading to compromised connective tissue integrity and function. This body of work has linked the biological aging of MSC to changes in their epigenetic signatures affecting growth, lifespan, self-renewal and multi-potential, due to deregulation of processes such as cellular senescence, oxidative stress, DNA damage, telomere shortening and DNA damage. This review addresses recent findings examining DNA methylation, histone modifications and miRNA changes in aging MSC populations. Moreover, we explore how epigenetic factors alter cellular pathways and associated biological networks, contributing to the MSC aging phenotype. Finally we discuss the crucial areas requiring a greater understanding of these processes, in order to piece together a global picture of the changing epigenetic landscape in MSC during aging.
Collapse
Affiliation(s)
- Dimitrios Cakouros
- Mesenchymal Stem Cell Laboratory, School of Medical Sciences, Faculty of Health Sciences, The University of Adelaide, Adelaide, SA, Australia; South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, School of Medical Sciences, Faculty of Health Sciences, The University of Adelaide, Adelaide, SA, Australia; South Australian Health and Medical Research Institute, Adelaide, SA, Australia.
| |
Collapse
|
153
|
Abstract
Over the past 100 y, tremendous progress has been made in the fields of dental tissue engineering and regenerative dental medicine, collectively known as translational dentistry. Translational dentistry has benefited from the more mature field of tissue engineering and regenerative medicine (TERM), established on the belief that biocompatible scaffolds, cells, and growth factors could be used to create functional, living replacement tissues and organs. TERM, created and pioneered by an interdisciplinary group of clinicians, biomedical engineers, and basic research scientists, works to create bioengineered replacement tissues that provide at least enough function for patients to survive until donor organs are available and, at best, fully functional replacement organs. Ultimately, the goal of both TERM and regenerative dentistry is to bring new and more effective therapies to the clinic to treat those in need. Very recently, the National Institutes of Health/National Institute of Dental and Craniofacial Research invested $24 million over a 3-y period to create dental oral and craniofacial translational resource centers to facilitate the development of more effective therapies to treat edentulism and other dental-related diseases over the next decade. This exciting era in regenerative dentistry, particularly for whole-tooth tissue engineering, builds on many key successes over the past 100 y that have contributed toward our current knowledge and understanding of signaling pathways directing natural tooth and dental tissue development-the foundation for current strategies to engineer functional, living replacement dental tissues and whole teeth. Here we use a historical perspective to present key findings and pivotal advances made in the field of translational dentistry over the past 100 y. We will first describe how this process has evolved over the past 100 y and then hypothesize on what to expect over the next century.
Collapse
Affiliation(s)
- P C Yelick
- Tufts University School of Dental Medicine, Division of Craniofacial and Molecular Genetics, Department of Orthodontics, Boston, MA, USA
| | - P T Sharpe
- Kings College London Dental Institute, London, UK
| |
Collapse
|
154
|
Costa LA, Eiro N, Fraile M, Gonzalez LO, Saá J, Garcia-Portabella P, Vega B, Schneider J, Vizoso FJ. Functional heterogeneity of mesenchymal stem cells from natural niches to culture conditions: implications for further clinical uses. Cell Mol Life Sci 2020; 78:447-467. [PMID: 32699947 PMCID: PMC7375036 DOI: 10.1007/s00018-020-03600-0] [Citation(s) in RCA: 195] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/02/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells (MSC) are present in all organs and tissues. Several studies have shown the therapeutic potential effect of MSC or their derived products. However, the functional heterogeneity of MSC constitutes an important barrier for transferring these capabilities to the clinic. MSC heterogeneity depends on their origin (biological niche) or the conditions of potential donors (age, diseases or unknown factors). It is accepted that many culture conditions of the artificial niche to which they are subjected, such as O2 tension, substrate and extracellular matrix cues, inflammatory stimuli or genetic manipulations can influence their resulting phenotype. Therefore, to attain a more personalized and precise medicine, a correct selection of MSC is mandatory, based on their functional potential, as well as the need to integrate all the existing information to achieve an optimal improvement of MSC features in the artificial niche.
Collapse
Affiliation(s)
- Luis A Costa
- Unidad de Investigación, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33920, Gijón, Asturias, Spain
| | - Noemi Eiro
- Unidad de Investigación, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33920, Gijón, Asturias, Spain
| | - María Fraile
- Unidad de Investigación, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33920, Gijón, Asturias, Spain
| | - Luis O Gonzalez
- Unidad de Investigación, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33920, Gijón, Asturias, Spain.,Department of Anatomical Pathology, Fundación Hospital de Jove, Gijón, Spain
| | - Jorge Saá
- Unidad de Investigación, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33920, Gijón, Asturias, Spain
| | - Pablo Garcia-Portabella
- Unidad de Investigación, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33920, Gijón, Asturias, Spain
| | - Belén Vega
- Unidad de Investigación, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33920, Gijón, Asturias, Spain
| | - José Schneider
- Department of Obstetrics and Gynecology, University of Valladolid, Valladolid, Spain
| | - Francisco J Vizoso
- Unidad de Investigación, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33920, Gijón, Asturias, Spain.
| |
Collapse
|
155
|
Sonoda S, Murata S, Nishida K, Kato H, Uehara N, Kyumoto YN, Yamaza H, Takahashi I, Kukita T, Yamaza T. Extracellular vesicles from deciduous pulp stem cells recover bone loss by regulating telomerase activity in an osteoporosis mouse model. Stem Cell Res Ther 2020; 11:296. [PMID: 32680564 PMCID: PMC7367365 DOI: 10.1186/s13287-020-01818-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/23/2020] [Accepted: 07/08/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Systemic transplantation of stem cells from human exfoliated deciduous teeth (SHED) recovers bone loss in animal models of osteoporosis; however, the mechanisms underlying this remain unclear. Here, we hypothesized that trophic factors within SHED-releasing extracellular vesicles (SHED-EVs) rescue osteoporotic phenotype. METHODS EVs were isolated from culture supernatant of SHED. SHED-EVs were treated with or without ribonuclease and systemically administrated into ovariectomized mice, followed by the function of recipient bone marrow mesenchymal stem cells (BMMSCs) including telomerase activity, osteoblast differentiation, and sepmaphorine-3A (SEMA3A) secretion. Subsequently, human BMMSCs were stimulated by SHED-EVs with or without ribonuclease treatment, and then human BMMSCs were examined regarding the function of telomerase activity, osteoblast differentiation, and SEMA3A secretion. Furthermore, SHED-EV-treated human BMMSCs were subcutaneously transplanted into the dorsal skin of immunocompromised mice with hydroxyapatite tricalcium phosphate (HA/TCP) careers and analyzed the de novo bone-forming ability. RESULTS We revealed that systemic SHED-EV-infusion recovered bone volume in ovariectomized mice and improved the function of recipient BMMSCs by rescuing the mRNA levels of Tert and telomerase activity, osteoblast differentiation, and SEMA3A secretion. Ribonuclease treatment depleted RNAs, including microRNAs, within SHED-EVs, and these RNA-depleted SHED-EVs attenuated SHED-EV-rescued function of recipient BMMSCs in the ovariectomized mice. These findings were supported by in vitro assays using human BMMSCs incubated with SHED-EVs. CONCLUSION Collectively, our findings suggest that SHED-secreted RNAs, such as microRNAs, play a crucial role in treating postmenopausal osteoporosis by targeting the telomerase activity of recipient BMMSCs.
Collapse
Affiliation(s)
- Soichiro Sonoda
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Sara Murata
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
- Section of Orthodontics and Dentofacial Orthopedics, Division of Oral Health, Growth & Development, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Kento Nishida
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hiroki Kato
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Norihisa Uehara
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yukari N Kyumoto
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Haruyoshi Yamaza
- Department of Pediatric Dentistry, Division of Oral Health, Growth & Development, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Ichiro Takahashi
- Section of Orthodontics and Dentofacial Orthopedics, Division of Oral Health, Growth & Development, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Toshio Kukita
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takayoshi Yamaza
- Department of Molecular Cell Biology and Oral Anatomy, Division of Oral Biological Sciences, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
156
|
Brzoska E, Kalkowski L, Kowalski K, Michalski P, Kowalczyk P, Mierzejewski B, Walczak P, Ciemerych MA, Janowski M. Muscular Contribution to Adolescent Idiopathic Scoliosis from the Perspective of Stem Cell-Based Regenerative Medicine. Stem Cells Dev 2020; 28:1059-1077. [PMID: 31170887 DOI: 10.1089/scd.2019.0073] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Adolescent idiopathic scoliosis (AIS) is a relatively frequent disease within a range 0.5%-5.0% of population, with higher frequency in females. While a resultant spinal deformity is usually medically benign condition, it produces far going psychosocial consequences, which warrant attention. The etiology of AIS is unknown and current therapeutic approaches are symptomatic only, and frequently inconvenient or invasive. Muscular contribution to AIS is widely recognized, although it did not translate to clinical routine as yet. Muscle asymmetry has been documented by pathological examinations as well as systemic muscle disorders frequently leading to scoliosis. It has been also reported numerous genetic, metabolic and radiological alterations in patients with AIS, which are linked to muscular and neuromuscular aspects. Therefore, muscles might be considered an attractive and still insufficiently exploited therapeutic target for AIS. Stem cell-based regenerative medicine is rapidly gaining momentum based on the tremendous progress in understanding of developmental biology. It comes also with a toolbox of various stem cells such as satellite cells or mesenchymal stem cells, which could be transplanted; also, the knowledge acquired in research on regenerative medicine can be applied to manipulation of endogenous stem cells to obtain desired therapeutic goals. Importantly, paravertebral muscles are located relatively superficially; therefore, they can be an easy target for minimally invasive approaches to treatment of AIS. It comes in pair with a fast progress in image guidance, which allows for precise delivery of therapeutic agents, including stem cells to various organs such as brain, muscles, and others. Summing up, it seems that there is a link between AIS, muscles, and stem cells, which might be worth of further investigations with a long-term goal of setting foundations for eventual bench-to-bedside translation.
Collapse
Affiliation(s)
- Edyta Brzoska
- 1Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Lukasz Kalkowski
- 2Department of Neurology and Neurosurgery, Faculty of Medical Sciences, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Kamil Kowalski
- 1Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Pawel Michalski
- 3Spine Surgery Department, Institute of Mother and Child, Warsaw, Poland
| | - Pawel Kowalczyk
- 4Department of Neurosurgery, Children's Memorial Health Institute, Warsaw, Poland
| | - Bartosz Mierzejewski
- 1Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Piotr Walczak
- 5Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,6Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Maria A Ciemerych
- 1Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Miroslaw Janowski
- 5Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,6Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
157
|
Habibalahi A, Moghari MD, Campbell JM, Anwer AG, Mahbub SB, Gosnell M, Saad S, Pollock C, Goldys EM. Non-invasive real-time imaging of reactive oxygen species (ROS) using auto-fluorescence multispectral imaging technique: A novel tool for redox biology. Redox Biol 2020; 34:101561. [PMID: 32526699 PMCID: PMC7287272 DOI: 10.1016/j.redox.2020.101561] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/23/2020] [Accepted: 04/27/2020] [Indexed: 02/06/2023] Open
Abstract
Detecting reactive oxygen species (ROS) that play a critical role as redox modulators and signalling molecules in biological systems currently requires invasive methods such as ROS -specific indicators for imaging and quantification. We developed a non-invasive, real-time, label-free imaging technique for assessing the level of ROS in live cells and thawed cryopreserved tissues that is compatible with in-vivo imaging. The technique is based on autofluorescence multispectral imaging (AFMI) carried out in an adapted fluorescence microscope with an expanded number of spectral channels spanning specific excitation (365 nm-495 nm) and emission (420 nm-700 nm) wavelength ranges. We established a strong quantitative correlation between the spectral information obtained from AFMI and the level of ROS obtained from CellROX staining. The results were obtained in several cell types (HeLa, PANC1 and mesenchymal stem cells) and in live kidney tissue. Additioanly,two spectral regimes were considered: with and without UV excitation (wavelengths > 400 nm); the latter being suitable for UV-sensitive systems such as the eye. Data were analyzed by linear regression combined with an optimization method of swarm intelligence. This allowed the calibration of AFMI signals to the level of ROS with excellent correlation (R = 0.84, p = 0.00) in the entire spectral range and very good correlation (R = 0.78, p = 0.00) in the limited, UV-free spectral range. We also developed a strong classifier which allowed us to distinguish moderate and high levels of ROS in these two regimes (AUC = 0.91 in the entire spectral range and AUC = 0.78 for UV-free imaging). These results indicate that ROS in cells and tissues can be imaged non-invasively, which opens the way to future clinical applications in conditions where reactive oxygen species are known to contribute to progressive disease such as in ophthalmology, diabetes, kidney disease, cancer and neurodegenerative diseases.
Collapse
Affiliation(s)
- Abbas Habibalahi
- ARC Centre of Excellence Centre for Nanoscale Biophotonics, University of New South Wales, Kensington, 2052, NSW, Australia.
| | - Mahdieh Dashtbani Moghari
- School of Biomedical Engineering, Faculty of Engineering, Darlington Campus, The University of Sydney, NSW, 2006, Australia
| | - Jared M Campbell
- ARC Centre of Excellence Centre for Nanoscale Biophotonics, University of New South Wales, Kensington, 2052, NSW, Australia
| | - Ayad G Anwer
- ARC Centre of Excellence Centre for Nanoscale Biophotonics, University of New South Wales, Kensington, 2052, NSW, Australia
| | - Saabah B Mahbub
- ARC Centre of Excellence Centre for Nanoscale Biophotonics, University of New South Wales, Kensington, 2052, NSW, Australia
| | | | - Sonia Saad
- Kolling Institute of Medical Research, University of Sydney, Camperdown, 2006, NSW, Australia
| | - Carol Pollock
- Kolling Institute of Medical Research, University of Sydney, Camperdown, 2006, NSW, Australia
| | - Ewa M Goldys
- ARC Centre of Excellence Centre for Nanoscale Biophotonics, University of New South Wales, Kensington, 2052, NSW, Australia
| |
Collapse
|
158
|
Cheng Q, Lin J, Chen Q, Zheng L, Tang Y, Wang F, Huang X, Zhang Y, Li S, Yang Z, Zhou P, He TC, Luo W, Zhang H. Role of Special AT-Rich Sequence-Binding Protein 2 in the Osteogenesis of Human Dental Mesenchymal Stem Cells. Stem Cells Dev 2020; 29:1059-1072. [PMID: 32484035 DOI: 10.1089/scd.2020.0013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Dental mesenchymal stem cells (MSCs) are recognized as a critical factor in repair of defective craniofacial bone owing to the multiple differentiation potential, the ability to regenerate distinct tissues, and the advantage that they can be easily obtained by relatively noninvasive procedures. Special AT-rich sequence-binding protein 2 (SATB2) is a nuclear matrix protein, involved in chromatin remodeling and transcriptional regulation, and has been reported to be as a positive regulator of osteoblast differentiation, bone formation, and bone regeneration in MSCs. In this study, we systematically investigated the capability of SATB2 to promote the osteogenic differentiation of periodontal ligament stem cells (PDLSCs), dental pulp stem cells (DPSCs), and stem cells from human exfoliated deciduous teeth (SHED). RNA-seq analysis and quantitative real-time PCR (RT-PCR) revealed that genes regulating osteogenic differentiation were differentially expressed among three cell types and SATB2 was found to be expressed at a relatively high level. When the three cell types overexpressed SATB2 with AdSATB2 infection, alkaline phosphatase (ALP) staining, ALP activity, Alizarin Red S staining, and quantification tended to increase with an increasing infection rate. It showed opposite results after infection with AdsiSATB2. RNA-seq analysis indicated that the expression of downstream osteogenic genes was affected by AdSATB2 infection and quantitative RT-PCR confirmed that nine osteogenic genes (Spp1, Sema7a, Atf4, Ibsp, Col1a1, Sp7, Igfbp3, Dlx3, and Alpl) were upregulated, to various extents, following SATB2 overexpression. In addition, quantitative PCR results indicated that SATB2 affected the expression of MSC markers. These results suggested an important role of SATB2 in the osteogenesis of PDLSCs, DPSCs, and SHED. Further research is warranted to investigate SATB2-mediated regulation of osteogenic differentiation and to evaluate the therapeutic use of SATB2 for the regeneration of defective craniofacial bone tissue.
Collapse
Affiliation(s)
- Qianyu Cheng
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China.,Department of Pediatric Dentistry, The Affiliated Stomatology Hospital, Chongqing Medical University, Chongqing, China
| | - Juhong Lin
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China.,Department of Pediatric Dentistry, The Affiliated Stomatology Hospital, Chongqing Medical University, Chongqing, China
| | - Qiuman Chen
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China.,Department of Pediatric Dentistry, The Affiliated Stomatology Hospital, Chongqing Medical University, Chongqing, China
| | - Liwen Zheng
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China.,Department of Pediatric Dentistry, The Affiliated Stomatology Hospital, Chongqing Medical University, Chongqing, China
| | - Yingying Tang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China.,Department of Pediatric Dentistry, The Affiliated Stomatology Hospital, Chongqing Medical University, Chongqing, China
| | - Feilong Wang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China.,Department of Pediatric Dentistry, The Affiliated Stomatology Hospital, Chongqing Medical University, Chongqing, China
| | - Xia Huang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Yuxin Zhang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China.,Department of Pediatric Dentistry, The Affiliated Stomatology Hospital, Chongqing Medical University, Chongqing, China
| | - Shuang Li
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China.,Department of Pediatric Dentistry, The Affiliated Stomatology Hospital, Chongqing Medical University, Chongqing, China
| | - Zhuohui Yang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China.,Department of Pediatric Dentistry, The Affiliated Stomatology Hospital, Chongqing Medical University, Chongqing, China
| | - Pengfei Zhou
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Tong-Chuan He
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China.,Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois, USA
| | - Wenping Luo
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Hongmei Zhang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China.,Department of Pediatric Dentistry, The Affiliated Stomatology Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
159
|
Tsai AC, Jeske R, Chen X, Yuan X, Li Y. Influence of Microenvironment on Mesenchymal Stem Cell Therapeutic Potency: From Planar Culture to Microcarriers. Front Bioeng Biotechnol 2020; 8:640. [PMID: 32671039 PMCID: PMC7327111 DOI: 10.3389/fbioe.2020.00640] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/26/2020] [Indexed: 12/15/2022] Open
Abstract
Human mesenchymal stem cells (hMSCs) are a promising candidate in cell therapy as they exhibit multilineage differentiation, homing to the site of injury, and secretion of trophic factors that facilitate tissue healing and/or modulate immune response. As a result, hMSC-derived products have attracted growing interests in preclinical and clinical studies. The development of hMSC culture platforms for large-scale biomanufacturing is necessary to meet the requirements for late-phase clinical trials and future commercialization. Microcarriers in stirred-tank bioreactors have been widely utilized in large-scale expansion of hMSCs for translational applications because of a high surface-to-volume ratio compared to conventional 2D planar culture. However, recent studies have demonstrated that microcarrier-expanded hMSCs differ from dish- or flask-expanded cells in size, morphology, proliferation, viability, surface markers, gene expression, differentiation potential, and secretome profile which may lead to altered therapeutic potency. Therefore, understanding the bioprocessing parameters that influence hMSC therapeutic efficacy is essential for the optimization of microcarrier-based bioreactor system to maximize hMSC quantity without sacrificing quality. In this review, biomanufacturing parameters encountered in planar culture and microcarrier-based bioreactor culture of hMSCs are compared and discussed with specific focus on cell-adhesion surface (e.g., discontinuous surface, underlying curvature, microcarrier stiffness, porosity, surface roughness, coating, and charge) and the dynamic microenvironment in bioreactor culture (e.g., oxygen and nutrients, shear stress, particle collision, and aggregation). The influence of dynamic culture in bioreactors on hMSC properties is also reviewed in order to establish connection between bioprocessing and stem cell function. This review addresses fundamental principles and concepts for future design of biomanufacturing systems for hMSC-based therapy.
Collapse
Affiliation(s)
- Ang-Chen Tsai
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| | - Richard Jeske
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| | - Xingchi Chen
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| | - Xuegang Yuan
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| |
Collapse
|
160
|
Comparison of skeletal and soft tissue pericytes identifies CXCR4 + bone forming mural cells in human tissues. Bone Res 2020; 8:22. [PMID: 32509378 PMCID: PMC7244476 DOI: 10.1038/s41413-020-0097-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 03/08/2020] [Accepted: 03/12/2020] [Indexed: 12/24/2022] Open
Abstract
Human osteogenic progenitors are not precisely defined, being primarily studied as heterogeneous multipotent cell populations and termed mesenchymal stem cells (MSCs). Notably, select human pericytes can develop into bone-forming osteoblasts. Here, we sought to define the differentiation potential of CD146+ human pericytes from skeletal and soft tissue sources, with the underlying goal of defining cell surface markers that typify an osteoblastogenic pericyte. CD146+CD31-CD45- pericytes were derived by fluorescence-activated cell sorting from human periosteum, adipose, or dermal tissue. Periosteal CD146+CD31-CD45- cells retained canonical features of pericytes/MSC. Periosteal pericytes demonstrated a striking tendency to undergo osteoblastogenesis in vitro and skeletogenesis in vivo, while soft tissue pericytes did not readily. Transcriptome analysis revealed higher CXCR4 signaling among periosteal pericytes in comparison to their soft tissue counterparts, and CXCR4 chemical inhibition abrogated ectopic ossification by periosteal pericytes. Conversely, enrichment of CXCR4+ pericytes or stromal cells identified an osteoblastic/non-adipocytic precursor cell. In sum, human skeletal and soft tissue pericytes differ in their basal abilities to form bone. Diversity exists in soft tissue pericytes, however, and CXCR4+ pericytes represent an osteoblastogenic, non-adipocytic cell precursor. Indeed, enrichment for CXCR4-expressing stromal cells is a potential new tactic for skeletal tissue engineering.
Collapse
|
161
|
Lavrentieva A, Hoffmann A, Lee-Thedieck C. Limited Potential or Unfavorable Manipulations? Strategies Toward Efficient Mesenchymal Stem/Stromal Cell Applications. Front Cell Dev Biol 2020; 8:316. [PMID: 32509777 PMCID: PMC7248306 DOI: 10.3389/fcell.2020.00316] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/09/2020] [Indexed: 02/06/2023] Open
Abstract
Despite almost 50 years of research and over 20 years of preclinical and clinical studies, the question of curative potential of mesenchymal stem/stromal cells (MSCs) is still widely discussed in the scientific community. Non-reproducible treatment outcomes or even absence of treatment effects in comparison to control groups challenges the potential of these cells for routine application both in tissue engineering and in regenerative medicine. One of the reasons of such outcomes is non-standardized and often disadvantageous ex vivo manipulation of MSCs prior therapy. In most cases, clinically relevant cell numbers for MSC-based therapies can be only obtained by in vitro expansion of isolated cells. In this mini review, we will discuss point by point possible pitfalls in the production of human MSCs for cell therapies, without consideration of material-based applications. Starting with cell source, choice of donor and recipient, as well as isolation methods, we will then discuss existing expansion protocols (two-/three-dimensional cultivation, basal medium, medium supplements, static/dynamic conditions, and hypoxic/normoxic conditions) and influence of these strategies on the cell functionality after implantation. The role of potency assays will also be addressed. The final aim of this mini review is to illustrate the heterogeneity of current strategies for gaining MSCs for clinical applications with their strengths and weaknesses. Only a careful consideration and standardization of all pretreatment processes/methods for the different applications of MSCs will ensure robust and reproducible performance of these cell populations in the different experimental and clinical settings.
Collapse
Affiliation(s)
| | - Andrea Hoffmann
- Department of Orthopaedic Surgery, Graded Implants and Regenerative Strategies, Hannover Medical School, Hanover, Germany
| | - Cornelia Lee-Thedieck
- Institute of Cell Biology and Biophysics, Leibniz University Hannover, Hanover, Germany
| |
Collapse
|
162
|
Behm C, Blufstein A, Gahn J, Nemec M, Moritz A, Rausch-Fan X, Andrukhov O. Cytokines Differently Define the Immunomodulation of Mesenchymal Stem Cells from the Periodontal Ligament. Cells 2020; 9:1222. [PMID: 32423044 PMCID: PMC7290931 DOI: 10.3390/cells9051222] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/05/2020] [Accepted: 05/11/2020] [Indexed: 12/11/2022] Open
Abstract
Human periodontal ligament stem cells (hPDLSCs) play an important role in periodontal tissue homeostasis and regeneration. The function of these cells in vivo depends largely on their immunomodulatory ability, which is reciprocally regulated by immune cells via cytokines, particularly interferon (IFN)-γ, tumor necrosis factor (TNF)-α, and interleukin (IL)-1β. Different cytokines activate distinct signaling pathways and might differently affect immunomodulatory activities of hPDLSCs. This study directly compared the effect of IFN-γ, TNF-α, or IL-1β treated primary hPDLSCs on allogenic CD4+ T lymphocyte proliferation and apoptosis in an indirect co-culture model. The effects of IFN-γ, TNF-α, and IL-1β on the expression of specific immunomodulatory factors such as intoleamine-2,3-dioxygenase-1 (IDO-1), prostaglandin E2 (PGE2), and programmed cell death 1 ligand 1 (PD-L1) and ligand 2 (PD-L2) in hPDLSCs were compared. The contribution of different immunomodulatory mediators to the immunomodulatory effects of hPDLSCs in the indirect co-culture experiments was assessed using specific inhibitors. Proliferation of CD4+ T lymphocytes was inhibited by hPDLSCs, and this effect was strongly enhanced by IFN-γ and IL-1β but not by TNF-α. Apoptosis of CD4+ T lymphocytes was decreased by hPDLSCs per se. This effect was counteracted by IFN-γ or IL-1β. Additionally, IFN-γ, TNF-α, and IL-1β differently regulated all investigated immunomediators in hPDLSCs. Pharmacological inhibition of immunomediators showed that their contribution in regulating CD4+ T lymphocytes depends on the cytokine milieu. Our data indicate that inflammatory cytokines activate specific immunomodulatory mechanisms in hPDLSCs and the expression of particular immunomodulatory factors, which underlies a complex reciprocal interaction between hPDLSCs and CD4+ T lymphocytes.
Collapse
Affiliation(s)
- Christian Behm
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (C.B.); (A.B.); (J.G.); (A.M.); (X.R.-F.)
- Department of Orthodontics, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria;
| | - Alice Blufstein
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (C.B.); (A.B.); (J.G.); (A.M.); (X.R.-F.)
| | - Johannes Gahn
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (C.B.); (A.B.); (J.G.); (A.M.); (X.R.-F.)
| | - Michael Nemec
- Department of Orthodontics, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria;
| | - Andreas Moritz
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (C.B.); (A.B.); (J.G.); (A.M.); (X.R.-F.)
| | - Xiaohui Rausch-Fan
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (C.B.); (A.B.); (J.G.); (A.M.); (X.R.-F.)
| | - Oleh Andrukhov
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria; (C.B.); (A.B.); (J.G.); (A.M.); (X.R.-F.)
| |
Collapse
|
163
|
Ueda T, Inden M, Ito T, Kurita H, Hozumi I. Characteristics and Therapeutic Potential of Dental Pulp Stem Cells on Neurodegenerative Diseases. Front Neurosci 2020; 14:407. [PMID: 32457568 PMCID: PMC7222959 DOI: 10.3389/fnins.2020.00407] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/03/2020] [Indexed: 12/13/2022] Open
Abstract
To evaluate the therapeutic potential of stem cells for neurodegenerative diseases, emphasis should be placed on clarifying the characteristics of the various types of stem cells. Among stem cells, dental pulp stem cells (DPSCs) are a cell population that is rich in cell proliferation and multipotency. It has been reported that transplantation of DPSCs has protective effects against models of neurodegenerative diseases. The protective effects are not only through differentiation into the target cell type for the disease but are also related to trophic factors released from DPSCs. Recently, it has been reported that serum-free culture supernatant of dental pulp stem cell-conditioned medium (DPCM) contains various trophic factors and cytokines and that DPCM is effective for models of neurodegenerative diseases such as Alzheimer’s disease (AD), Parkinson’s disease (PD), and Amyotrophic Lateral Sclerosis (ALS). Moreover, the use of stem cells from human exfoliated deciduous teeth (SHEDs) has been considered. SHEDs are derived from deciduous teeth that have been disposed of as medical waste. SHEDs have higher differentiation capacity and proliferation ability than DPSCs. In addition, the serum-free culture supernatant of SHEDs (SHED-CM) contains more trophic factors, cytokines, and biometals than DPCM and also promotes neuroprotection. The neuroprotective effect of DPSCs, including those from deciduous teeth, will be used as the seeds of therapeutic drugs for neurodegenerative diseases. SHEDs will be used for further cell therapy of neurodegenerative diseases in the future. In this paper, we focused on the characteristics of DPSCs and their potential for neurodegenerative diseases.
Collapse
Affiliation(s)
- Tomoyuki Ueda
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Masatoshi Inden
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Taisei Ito
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Hisaka Kurita
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Isao Hozumi
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
164
|
Gopinath VK, Soumya S, Jayakumar MN. Osteogenic and odontogenic differentiation potential of dental pulp stem cells isolated from inflamed dental pulp tissues (I-DPSCs) by two different methods. Acta Odontol Scand 2020; 78:281-289. [PMID: 31855089 DOI: 10.1080/00016357.2019.1702716] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Objective: The objective of the present study is to isolate stem cells from inflamed dental pulp tissues (I-DPSCs) and study the characteristic such as surface markers, osteo/odontogenic differentiation potential between the outgrowth (OG) and enzymatic digestion (COL) methods.Materials and methods: I-DPSCs harvested by both methods were analysed for Mesenchymal Stem Cell marker expression by flow cytometry. The metabolic activity of the isolated cells was assessed by MTT assay. The Alkaline Phosphatase (ALP) and Alizarin red staining was done to analyse the osteogenic potential of isolated cells. The osteo/odontogenic differentiation was done by checking the expression of Dentine Matrix Protein 1 (DMP1), Dentine Sialophosphoprotein (DSPP), ALP and Bone Gamma-Carboxyglutamate Protein (BGLAP) by Real time PCR.Results: The isolated cells were positive for MSC markers such as CD-90, CD-105 and CD-73 and negative for CD-14, CD-45 and STRO-1. MTT assay indicated that the I-DPSCs from OG method showed higher metabolic activity than cells from COL. However, the osteo/odontogenic differentiation was in favour of cells isolated by COL method.Conclusion: Although the cell metabolic rate was more in OG, the osteo/odontogenic differentiation was higher in COL, suggesting that the isolation method and culture conditions do affect the differentiation capacity of isolated cells.
Collapse
Affiliation(s)
- Vellore Kannan Gopinath
- Department of Preventive and Restorative Dentistry, College of Dental Medicine, University of Sharjah, Sharjah, UAE
| | - S. Soumya
- The Sharjah Institute for Medical Research, University of Sharjah, Sharjah, UAE
| | | |
Collapse
|
165
|
Yianni V, Sharpe PT. Epigenetic mechanisms driving lineage commitment in mesenchymal stem cells. Bone 2020; 134:115309. [PMID: 32145460 DOI: 10.1016/j.bone.2020.115309] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/02/2020] [Accepted: 03/02/2020] [Indexed: 12/15/2022]
Abstract
The increasing application of approaches that allow tracing of individual cells over time, together with transcriptomic and epigenomic analyses is changing the way resident stromal stem cells (mesenchymal stem cells) are viewed. Rather than being a defined, homogeneous cell population as described following in vitro expansion, in vivo, these cells are highly programmed according to their resident tissue location. This programming is evidenced by different epigenetic landscapes and gene transcription signatures in cells before any in vitro expansion. This has potentially profound implications for the heterotypic use of these cells in therapeutic tissue engineering applications.
Collapse
Affiliation(s)
- Val Yianni
- Centre for Craniofacial & Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, United Kingdom of Great Britain and Northern Ireland
| | - Paul T Sharpe
- Centre for Craniofacial & Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, United Kingdom of Great Britain and Northern Ireland.
| |
Collapse
|
166
|
Rahayu RP, Pribadi N, Widjiastuti I, Nugrahani NA. Combinations of propolis and Ca(OH)2 in dental pulp capping treatment for the stimulation of reparative dentin formation in a rat model. F1000Res 2020; 9:308. [PMID: 32733674 PMCID: PMC7369428 DOI: 10.12688/f1000research.22409.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/17/2020] [Indexed: 11/20/2022] Open
Abstract
Background: Caries in the dental pulp result in inflammation and damage to the pulp tissue. During inflammation of the pulp, various inflammatory mediators and growth factors are released, including IL-8, IL-10, TLR-2, VEGF and TGF-β through the NF-kB pathway. In the present study, therapy for pulpal caries was performed through pulp capping by giving a combination of propolis and calcium hydroxide (Ca(OH)2). This treatment was expected to stimulate the formation of reparative dentin as an anti-inflammatory material to prevent pulp tissue damage. Methods: 28 Wistar rats were divided into four groups and treated with Ca(OH)2 with or without the addition of propolis for either 7 or 14 days. Immunohistochemical examination was used to determine the expression of IL-8, IL-10, TLR-2, VEGF, TGF-β in the four treatment groups. Results: The group treated with a combination of propolis and Ca(OH)2 for 7 days showed that the expression of IL-10, IL-8, TLR-2, VEGF, TGF-β increased significantly compared to the treatment group treated with only Ca(OH)2. The expression of IL-10, TLR-2, TGF-β, VEGF increased in the treatment group treated with propolis and Ca(OH)2 for 14 days, while the expression of IL-8 in the decreased significantly. Conclusions: Administration of a combination of propolis and Ca(OH)2 has efficacy in the pulp capping treatment process because it has anti-bacterial and immunomodulatory properties. The results show that it is able to stimulate the process of pulp tissue repair through increased expression of IL-10, TGF-β, VEGF, TLR -2 and decreased expression of IL-8.
Collapse
Affiliation(s)
- Retno Pudji Rahayu
- Department of Oral and Maxillofacial Pathology, Faculty of Dentistry, Airlangga University, Surabaya, East Java, 60131, Indonesia
| | - Nirawati Pribadi
- Department of Conservative Dentistry, Faculty of Dentistry, Airlangga University, Surabaya, East Java, 60131, Indonesia
| | - Ira Widjiastuti
- Department of Conservative Dentistry, Faculty of Dentistry, Airlangga University, Surabaya, East Java, 60131, Indonesia
| | - Nur Ariska Nugrahani
- Immunology Study Program, Airlangga University, Surabaya, East Java, 60131, Indonesia
| |
Collapse
|
167
|
Lo Monaco M, Gervois P, Beaumont J, Clegg P, Bronckaers A, Vandeweerd JM, Lambrichts I. Therapeutic Potential of Dental Pulp Stem Cells and Leukocyte- and Platelet-Rich Fibrin for Osteoarthritis. Cells 2020; 9:cells9040980. [PMID: 32326610 PMCID: PMC7227024 DOI: 10.3390/cells9040980] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/07/2020] [Accepted: 04/13/2020] [Indexed: 12/18/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative and inflammatory joint disorder with cartilage loss. Dental pulp stem cells (DPSCs) can undergo chondrogenic differentiation and secrete growth factors associated with tissue repair and immunomodulation. Leukocyte- and platelet-rich fibrin (L-PRF) emerges in regenerative medicine because of its growth factor content and fibrin matrix. This study evaluates the therapeutic application of DPSCs and L-PRF in OA via immunomodulation and cartilage regeneration. Chondrogenic differentiation of DPSCs, with or without L-PRF exudate (ex) and conditioned medium (CM), and of bone marrow-mesenchymal stem cells was compared. These cells showed differential chondrogenesis. L-PRF was unable to increase cartilage-associated components. Immature murine articular chondrocytes (iMACs) were cultured with L-PRF ex, L-PRF CM, or DPSC CM. L-PRF CM had pro-survival and proliferative effects on unstimulated and cytokine-stimulated iMACs. L-PRF CM stimulated the release of IL-6 and PGE2, and increased MMP-13, TIMP-1 and IL-6 mRNA levels in cytokine-stimulated iMACs. DPSC CM increased the survival and proliferation of unstimulated iMACs. In cytokine-stimulated iMACs, DPSC CM increased TIMP-1 gene expression, whereas it inhibited nitrite release in 3D culture. We showed promising effects of DPSCs in an in vitro OA model, as they undergo chondrogenesis in vitro, stimulate the survival of chondrocytes and have immunomodulatory effects.
Collapse
Affiliation(s)
- Melissa Lo Monaco
- Cardio & Organ Systems (COST), Biomedical Research Institute (BIOMED), Hasselt University, 3590 Diepenbeek, Belgium; (P.G.); (J.B.); (A.B.); (I.L.)
- Department of Veterinary Medicine, Integrated Veterinary Research Unit (IVRU) - Namur Research Institute for Life Science (NARILIS), University of Namur, 5000 Namur, Belgium;
- Correspondence: ; Tel.: +32-(0)-26-92-09
| | - Pascal Gervois
- Cardio & Organ Systems (COST), Biomedical Research Institute (BIOMED), Hasselt University, 3590 Diepenbeek, Belgium; (P.G.); (J.B.); (A.B.); (I.L.)
| | - Joel Beaumont
- Cardio & Organ Systems (COST), Biomedical Research Institute (BIOMED), Hasselt University, 3590 Diepenbeek, Belgium; (P.G.); (J.B.); (A.B.); (I.L.)
- Maastricht Radiation Oncology (MaastRO) Lab, GROW—School for Oncology and Developmental Biology, Maastricht University, 6229ER Maastricht, The Netherlands
| | - Peter Clegg
- Department of Musculoskeletal and Ageing Sciences, Institute of Lifecourse and Medical Sciences, University of Liverpool, L7 8TX Liverpool, UK;
| | - Annelies Bronckaers
- Cardio & Organ Systems (COST), Biomedical Research Institute (BIOMED), Hasselt University, 3590 Diepenbeek, Belgium; (P.G.); (J.B.); (A.B.); (I.L.)
| | - Jean-Michel Vandeweerd
- Department of Veterinary Medicine, Integrated Veterinary Research Unit (IVRU) - Namur Research Institute for Life Science (NARILIS), University of Namur, 5000 Namur, Belgium;
| | - Ivo Lambrichts
- Cardio & Organ Systems (COST), Biomedical Research Institute (BIOMED), Hasselt University, 3590 Diepenbeek, Belgium; (P.G.); (J.B.); (A.B.); (I.L.)
| |
Collapse
|
168
|
Makarevich PI, Efimenko AY, Tkachuk VA. Biochemical Regulation of Regenerative Processes by Growth Factors and Cytokines: Basic Mechanisms and Relevance for Regenerative Medicine. BIOCHEMISTRY (MOSCOW) 2020; 85:11-26. [PMID: 32079514 DOI: 10.1134/s0006297920010022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Regenerative medicine that had emerged as a scientific and medical discipline at end of 20th century uses cultured cells and tissue-engineered structures for transplantation into human body to restore lost or damaged organs. However, practical achievements in this field are far from the promising results obtained in laboratory experiments. Searching for new directions has made apparent that successful solution of practical problems is impossible without understanding the fundamental principles of the regulation of development, renewal, and regeneration of human tissues. These aspects have been extensively investigated by cell biologists, physiologists, and biochemists working in a specific research area often referred to as regenerative biology. It is known that during regeneration, growth factors, cytokines, and hormones act beyond the regulation of individual cell functions, but rather activate specific receptor systems and control pivotal tissue repair processes, including cell proliferation and differentiation. These events require numerous coordinated stimuli and, therefore, are practically irreproducible using single proteins or low-molecular-weight compounds, i.e., cannot be directed by applying classical pharmacological approaches. Our review summarizes current concepts on the regulatory mechanisms of renewal and regeneration of human tissues with special attention to certain general biological and evolutionary aspects. We focus on the biochemical regulatory mechanisms of regeneration, in particular, the role of growth factors and cytokines and their receptor systems. In a separate section, we discussed practical approaches for activating regeneration using small molecules and stem cell secretome containing a broad repertoire of growth factors, cytokines, peptides, and extracellular vesicles.
Collapse
Affiliation(s)
- P I Makarevich
- Lomonosov Moscow State University, Institute for Regenerative Medicine, Medical Research and Education Center, Moscow, 119991, Russia. .,Lomonosov Moscow State University, Faculty of Fundamental Medicine, Moscow, 119991, Russia
| | - A Yu Efimenko
- Lomonosov Moscow State University, Institute for Regenerative Medicine, Medical Research and Education Center, Moscow, 119991, Russia.,Lomonosov Moscow State University, Faculty of Fundamental Medicine, Moscow, 119991, Russia
| | - V A Tkachuk
- Lomonosov Moscow State University, Institute for Regenerative Medicine, Medical Research and Education Center, Moscow, 119991, Russia.,Lomonosov Moscow State University, Faculty of Fundamental Medicine, Moscow, 119991, Russia.,Institute of Experimental Cardiology, National Medical Research Center of Cardiology, Moscow, 121552, Russia
| |
Collapse
|
169
|
Zeitlin BD. Banking on teeth - Stem cells and the dental office. Biomed J 2020; 43:124-133. [PMID: 32381462 PMCID: PMC7283549 DOI: 10.1016/j.bj.2020.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/29/2020] [Accepted: 02/12/2020] [Indexed: 12/23/2022] Open
Abstract
Science and commerce advance together and the stem cell field is no exception. With the promise of cures for conditions as diverse as cancer, autism, neural degeneration, organ replacement and addiction, long-term preservation of dental stem cells is a growth market. The discovery nearly twenty years ago, of viable, multipotent, stem cells in dental pulp from both baby and adult teeth initiated, and drives, this market.The dental stem cell preservation services, "tooth banks", focus on the collection of a child's baby teeth, as they are shed naturally, and storage of the stem cells from within the pulp for therapeutic use in later years should the child require them. This review focuses on the procedures related to these stem cell storage services and may serve as an introduction for many to the practice of "tooth banking".
Collapse
Affiliation(s)
- Benjamin D Zeitlin
- University of the Pacific, Arthur A. Dugoni School of Dentistry, San Francisco, CA, USA.
| |
Collapse
|
170
|
Shi X, Mao J, Liu Y. Pulp stem cells derived from human permanent and deciduous teeth: Biological characteristics and therapeutic applications. Stem Cells Transl Med 2020; 9:445-464. [PMID: 31943813 PMCID: PMC7103623 DOI: 10.1002/sctm.19-0398] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 12/27/2019] [Indexed: 12/13/2022] Open
Abstract
Human pulp stem cells (PSCs) include dental pulp stem cells (DPSCs) isolated from dental pulp tissues of human extracted permanent teeth and stem cells from human exfoliated deciduous teeth (SHED). Depending on their multipotency and sensitivity to local paracrine activity, DPSCs and SHED exert therapeutic applications at multiple levels beyond the scope of the stomatognathic system. This review is specifically concentrated on PSC-updated biological characteristics and their promising therapeutic applications in (pre)clinical practice. Biologically, distinguished from conventional mesenchymal stem cell markers in vitro, NG2, Gli1, and Celsr1 have been evidenced as PSC markers in vivo. Both perivascular cells and glial cells account for PSC origin. Therapeutically, endodontic regeneration is where PSCs hold the most promises, attributable of PSCs' robust angiogenic, neurogenic, and odontogenic capabilities. More recently, the interplay between cell homing and liberated growth factors from dentin matrix has endowed a novel approach for pulp-dentin complex regeneration. In addition, PSC transplantation for extraoral tissue repair and regeneration has achieved immense progress, following their multipotential differentiation and paracrine mechanism. Accordingly, PSC banking is undergoing extensively with the intent of advancing tissue engineering, disease remodeling, and (pre)clinical treatments.
Collapse
Affiliation(s)
- Xin Shi
- Center of Stomatology, Tongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
| | - Jing Mao
- Center of Stomatology, Tongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
| | - Yan Liu
- Laboratory of Biomimetic Nanomaterials, Department of OrthodonticsPeking University School and Hospital of StomatologyBeijingPeople's Republic of China
| |
Collapse
|
171
|
Tsutsui TW. Dental Pulp Stem Cells: Advances to Applications. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2020; 13:33-42. [PMID: 32104005 PMCID: PMC7025818 DOI: 10.2147/sccaa.s166759] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 12/07/2019] [Indexed: 12/18/2022]
Abstract
Dental pulp stem cells (DPSCs) have a high capacity for differentiation and the ability to regenerate a dentin/pulp-like complex. Numerous studies have provided evidence of DPSCs’ differentiation capacity, such as in neurogenesis, adipogenesis, osteogenesis, chondrogenesis, angiogenesis, and dentinogenesis. The molecular mechanisms and functions of DPSCs’ differentiation process are affected by growth factors and scaffolds. For example, growth factors such as basic fibroblast growth factor (bFGF), transforming growth factor-β (TGF-β), nerve growth factor (NGF), platelet-derived growth factor (PDGF), and bone morphogenic proteins (BMPs) influence DPSC fate, including in differentiation, cell proliferation, and wound healing. In addition, several types of scaffolds, such as collagen, hydrogel, decellularized bioscaffold, and nanofibrous spongy microspheres, have been used to characterize DPSC cellular attachment, migration, proliferation, differentiation, and functions. An appropriate combination of growth factors and scaffolds can enhance the differentiation capacity of DPSCs, in terms of optimizing not only dental-related expression but also dental pulp morphology. For a cell-based clinical approach, focus has been placed on the tissue engineering triad [cells/bioactive molecules (growth factors)/scaffolds] to characterize DPSCs. It is clear that a deep understanding of the mechanisms of stem cells, including their aging, self-renewal, microenvironmental homeostasis, and differentiation correlated with cell activity, the energy for which is provided from mitochondria, should provide new approaches for DPSC research and therapeutics. Mitochondrial functions and dynamics are related to the direction of stem cell differentiation, including glycolysis, oxidative phosphorylation, mitochondrial metabolism, mitochondrial transcription factor A (TFAM), mitochondrial elongation, and mitochondrial fusion and fission proteins. This review summarizes the effects of major growth factors and scaffolds for regenerating dentin/pulp-like complexes, as well as elucidating mitochondrial properties of DPSCs for the development of advanced applications research.
Collapse
Affiliation(s)
- Takeo W Tsutsui
- Department of Pharmacology, School of Life Dentistry at Tokyo, The Nippon Dental University, Tokyo, Japan
| |
Collapse
|
172
|
Andrejew R, Glaser T, Oliveira-Giacomelli Á, Ribeiro D, Godoy M, Granato A, Ulrich H. Targeting Purinergic Signaling and Cell Therapy in Cardiovascular and Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1201:275-353. [PMID: 31898792 DOI: 10.1007/978-3-030-31206-0_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Extracellular purines exert several functions in physiological and pathophysiological mechanisms. ATP acts through P2 receptors as a neurotransmitter and neuromodulator and modulates heart contractility, while adenosine participates in neurotransmission, blood pressure, and many other mechanisms. Because of their capability to differentiate into mature cell types, they provide a unique therapeutic strategy for regenerating damaged tissue, such as in cardiovascular and neurodegenerative diseases. Purinergic signaling is pivotal for controlling stem cell differentiation and phenotype determination. Proliferation, differentiation, and apoptosis of stem cells of various origins are regulated by purinergic receptors. In this chapter, we selected neurodegenerative and cardiovascular diseases with clinical trials using cell therapy and purinergic receptor targeting. We discuss these approaches as therapeutic alternatives to neurodegenerative and cardiovascular diseases. For instance, promising results were demonstrated in the utilization of mesenchymal stem cells and bone marrow mononuclear cells in vascular regeneration. Regarding neurodegenerative diseases, in general, P2X7 and A2A receptors mostly worsen the degenerative state. Stem cell-based therapy, mainly through mesenchymal and hematopoietic stem cells, showed promising results in improving symptoms caused by neurodegeneration. We propose that purinergic receptor activity regulation combined with stem cells could enhance proliferative and differentiation rates as well as cell engraftment.
Collapse
Affiliation(s)
- Roberta Andrejew
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Talita Glaser
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Ágatha Oliveira-Giacomelli
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Deidiane Ribeiro
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Mariana Godoy
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil.,Laboratory of Neurodegenerative Diseases, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandro Granato
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Henning Ulrich
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
173
|
Yamauchi Y, Cooper PR, Shimizu E, Kobayashi Y, Smith AJ, Duncan HF. Histone Acetylation as a Regenerative Target in the Dentine-Pulp Complex. Front Genet 2020; 11:1. [PMID: 32117431 PMCID: PMC7016267 DOI: 10.3389/fgene.2020.00001] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 01/06/2020] [Indexed: 01/09/2023] Open
Abstract
If dental caries (or tooth decay) progresses without intervention, the infection will advance through the dentine leading to severe pulpal inflammation (irreversible pulpitis) and pulp death. The current management of irreversible pulpits is generally root-canal-treatment (RCT), a destructive, expensive, and often unnecessary procedure, as removal of the injurious stimulus alone creates an environment in which pulp regeneration may be possible. Current dental-restorative-materials stimulate repair non-specifically and have practical limitations; as a result, opportunities exist for the development of novel therapeutic strategies to regenerate the damaged dentine-pulp complex. Recently, epigenetic modification of DNA-associated histone ‘tails’ has been demonstrated to regulate the self-renewal and differentiation potential of dental-stem-cell (DSC) populations central to regenerative endodontic treatments. As a result, the activities of histone deacetylases (HDAC) are being recognised as important regulators of mineralisation in both tooth development and dental-pulp-repair processes, with HDAC-inhibition (HDACi) promoting pulp cell mineralisation in vitro and in vivo. Low concentration HDACi-application can promote de-differentiation of DSC populations and conversely, increase differentiation and accelerate mineralisation in DSC populations. Therapeutically, various HDACi solutions can release bioactive dentine-matrix-components (DMCs) from the tooth’s extracellular matrix; solubilised DMCs are rich in growth factors and can stimulate regenerative processes such as angiogenesis, neurogenesis, and mineralisation. The aim of this mini-review is to discuss the role of histone-acetylation in the regulation of DSC populations, while highlighting the importance of HDAC in tooth development and dental pulp regenerative-mineralisation processes, before considering the potential therapeutic application of HDACi in targeted biomaterials to the damaged pulp to stimulate regeneration.
Collapse
Affiliation(s)
- Yukako Yamauchi
- Division of Restorative Dentistry & Periodontology, Dublin Dental University Hospital, Trinity College Dublin, University of Dublin, Dublin, Ireland
| | - Paul Roy Cooper
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Emi Shimizu
- Oral Biology Department, Rutgers School of Dental Medicine, Newark, NJ, United States
| | - Yoshifumi Kobayashi
- Oral Biology Department, Rutgers School of Dental Medicine, Newark, NJ, United States
| | - Anthony J Smith
- Oral Biology, School of Dentistry, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Henry Fergus Duncan
- Division of Restorative Dentistry & Periodontology, Dublin Dental University Hospital, Trinity College Dublin, University of Dublin, Dublin, Ireland
| |
Collapse
|
174
|
Eph/ephrin Signaling and Biology of Mesenchymal Stromal/Stem Cells. J Clin Med 2020; 9:jcm9020310. [PMID: 31979096 PMCID: PMC7074403 DOI: 10.3390/jcm9020310] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/14/2020] [Accepted: 01/19/2020] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) have emerged as important therapeutic agents, owing to their easy isolation and culture, and their remarkable immunomodulatory and anti-inflammatory properties. However, MSCs constitute a heterogeneous cell population which does not express specific cell markers and has important problems for in vivo homing, and factors regulating their survival, proliferation, and differentiation are largely unknown. Accordingly, in the present article, we review the current evidence on the relationships between Eph kinase receptors, their ephrin ligands, and MSCs. These molecules are involved in the adult homeostasis of numerous tissues, and we and other authors have demonstrated their expression in human and murine MSCs derived from both bone marrow and adipose tissue, as well as their involvement in the MSC biology. We extend these studies providing new results on the effects of Eph/ephrins in the differentiation and immunomodulatory properties of MSCs.
Collapse
|
175
|
Ceccariglia S, Cargnoni A, Silini AR, Parolini O. Autophagy: a potential key contributor to the therapeutic action of mesenchymal stem cells. Autophagy 2020; 16:28-37. [PMID: 31185790 PMCID: PMC6984485 DOI: 10.1080/15548627.2019.1630223] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 06/04/2019] [Accepted: 06/04/2019] [Indexed: 12/19/2022] Open
Abstract
Macroautophagy/autophagy occurs at basal levels in all eukaryotic cells and plays an important role in maintaining bio-energetic homeostasis through the control of molecule degradation and organelle turnover. It can be induced by environmental conditions such as starvation, and is deregulated in many diseases including autoimmune diseases, neurodegenerative disorders, and cancer. Interestingly, the modulation of autophagy in mesenchymal stem cells (MSCs) represents a possible mechanism which, affecting MSC properties, may have an impact on their regenerative, therapeutic potential. Furthermore, the ability of MSCs to modulate autophagy of cells in injured tissues/organs has been recently proposed to be involved in the regeneration of damaged tissues and organs. In particular, MSCs can affect autophagy in immune cells involved in injury-induced inflammation reducing their survival, proliferation, and function and favoring the resolution of inflammation. In addition, MSCs can affect autophagy in endogenous adult or progenitor cells, promoting their survival, proliferation and differentiation supporting the restoration of functional tissue. This review provides, for the first time, an overview of the studies which highlight a possible link between the therapeutic properties of MSCs and their ability to modulate autophagy, and it summarizes examples of disorders where these therapeutic properties have been correlated with such modulation. A better elucidation of the mechanism(s) through which MSCs can modulate the autophagy of target cells and how autophagy can affect MSCs therapeutic properties, can provide a wider perspective for the clinical application of MSCs in the treatment of many diseases.Abbreviations: 3-MA: 3-methyladenine; AD: Alzheimer disease; ATG: autophagy-related; BECN1: beclin 1; BM: bone marrow; CD: cluster of differentiation; EAE: experimental autoimmune encephalomyelitis; IL: interleukin; INF: interferon; LAP: LC3-associated phagocytosis; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MSCs: mesenchymal stem cells; MTOR: mechanistic target of rapamycin kinase; PD: Parkinson disease; PtdIns3K: class III phosphatidylinositol 3-kinase; ROS: reactive oxygen species; SLE: systemic lupus erythematosus; SQSTM1: sequestosome 1; TBI: traumatic brain injury; TGF: transforming growth factor; TNF: tumor necrosis factor.
Collapse
Affiliation(s)
- Sabrina Ceccariglia
- Istituto di Anatomia Umana e Biologia Cellulare, Università Cattolica del Sacro Cuore, Roma, Italia
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| | - Anna Cargnoni
- Centro di Ricerca “E. Menni”, Fondazione Poliambulanza - Istituto Ospedaliero, Brescia, Italy
| | - Antonietta Rosa Silini
- Centro di Ricerca “E. Menni”, Fondazione Poliambulanza - Istituto Ospedaliero, Brescia, Italy
| | - Ornella Parolini
- Istituto di Anatomia Umana e Biologia Cellulare, Università Cattolica del Sacro Cuore, Roma, Italia
- Centro di Ricerca “E. Menni”, Fondazione Poliambulanza - Istituto Ospedaliero, Brescia, Italy
| |
Collapse
|
176
|
Behm C, Blufstein A, Gahn J, Kubin B, Nemec M, Moritz A, Rausch-Fan X, Andrukhov O. 1,25(OH) 2D 3 Differently Affects Immunomodulatory Activities of Mesenchymal Stem Cells Depending on the Presence of TNF-α, IL-1β and IFN-γ. J Clin Med 2019; 8:2211. [PMID: 31847340 PMCID: PMC6947512 DOI: 10.3390/jcm8122211] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 12/10/2019] [Accepted: 12/12/2019] [Indexed: 12/15/2022] Open
Abstract
Periodontal ligament-derived mesenchymal stem cells (hPDLSCs) possess immunomodulatory abilities which are strongly enhanced by various inflammatory cytokines. Vitamin D3 has anti-inflammatory effects on hPDLSCs and immune cells. However, no study to date has directly compared the influence of 1,25(OH)2D3 on the immunomodulatory activities of hPDLSCs in the presence of different cytokines. In the present study, the effects of hPDLSCs treated with tumor necrosis factor (TNF)-α, interleukin (IL)-1β, or interferon (IFN)-γ in the presence of 1,25(OH)2D3 on the proliferation of allogenic CD4+ T lymphocyte or on the functional status of primary CD68+ macrophages were analyzed in coculture models. Additionally, the effects of 1,25(OH)2D3 on TNF-α-, IL-1β-, and IFN-γ-induced gene expression of some immunomodulatory factors in hPDLSCs were compared. Under coculture conditions, 1,25(OH)2D3 increased or decreased CD4+ T lymphocyte proliferation via hPDLSCs, depending on the cytokine. hPDLSCs primed with 1,25(OH)2D3 and different cytokines affected pro- and anti-inflammatory cytokine expression in macrophages variably, depending on the priming cytokine. With one exception, 1,25(OH)2D3 significantly reduced TNF-α-, IL-1β-, and IFN-γ-induced expression of all the investigated immunomediators in hPDLSCs, albeit to different extents. These results suggest that 1,25(OH)2D3 influences the immunomodulatory activities of hPDLSCs depending qualitatively and quantitatively on the presence of certain inflammatory cytokines.
Collapse
Affiliation(s)
- Christian Behm
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, 1090 Wien, Austria; (C.B.); (A.B.); (J.G.); (B.K.); (A.M.); (X.R.-F.)
| | - Alice Blufstein
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, 1090 Wien, Austria; (C.B.); (A.B.); (J.G.); (B.K.); (A.M.); (X.R.-F.)
| | - Johannes Gahn
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, 1090 Wien, Austria; (C.B.); (A.B.); (J.G.); (B.K.); (A.M.); (X.R.-F.)
| | - Barbara Kubin
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, 1090 Wien, Austria; (C.B.); (A.B.); (J.G.); (B.K.); (A.M.); (X.R.-F.)
| | - Michael Nemec
- Clinical Division of Orthodontics, University Clinic of Dentistry, Medical University of Vienna, 1090 Wien, Austria;
| | - Andreas Moritz
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, 1090 Wien, Austria; (C.B.); (A.B.); (J.G.); (B.K.); (A.M.); (X.R.-F.)
| | - Xiaohui Rausch-Fan
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, 1090 Wien, Austria; (C.B.); (A.B.); (J.G.); (B.K.); (A.M.); (X.R.-F.)
| | - Oleh Andrukhov
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, 1090 Wien, Austria; (C.B.); (A.B.); (J.G.); (B.K.); (A.M.); (X.R.-F.)
| |
Collapse
|
177
|
Park MK, Kim S, Jeon M, Jung UW, Lee JH, Choi HJ, Choi JE, Song JS. Evaluation of the Apical Complex and the Coronal Pulp as a Stem Cell Source for Dentin-pulp Regeneration. J Endod 2019; 46:224-231.e3. [PMID: 31836138 DOI: 10.1016/j.joen.2019.10.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 01/09/2023]
Abstract
INTRODUCTION This study compared the stemness and differentiation potential of stem cells derived from the apical complex (apical complex cells [ACCs]) and coronal pulp (dental pulp stem cells [DPSCs]) of human immature permanent teeth with the aim of determining a more suitable source of stem cells for regeneration of the dentin-pulp complex. METHODS ACC and DPSC cultures were established from 13 human immature permanent teeth using the outgrowth method. The proliferation capacity and colony-forming ability of ACCs and DPSCs were evaluated. ACCs and DPSCs were analyzed for mesenchymal stem cell markers using flow cytometry. The adipogenic and osteogenic differentiation potential of ACCs and DPSCs were evaluated using the quantitative real-time polymerase chain reaction and histochemical staining. ACCs and DPSCs were transplanted subcutaneously in immunocompromised mice using macroporous biphasic calcium phosphate as a carrier. The histomorphologic characteristics of the newly formed tissues were verified using hematoxylin-eosin staining and immunohistochemical staining. Quantitative alkaline phosphatase analysis and quantitative real-time polymerase chain reaction using BSP, DSPP, POSTN, and ColXII were performed. RESULTS ACCs and DPSCs showed similar cell proliferation potential and colony-forming ability. The percentage of mesenchymal stem cell markers was similar between ACCs and DPSCs. In the in vitro study, ACCs and DPSCs showed adipogenic and osteogenic differentiation potential. In the in vivo study, ACCs and DPSCs formed amorphous hard tissue using macroporous biphasic calcium phosphate particles. The quantity and histomorphologic characteristics of the amorphous hard tissue were similar in the ACC and DPSC groups. Formation of periodontal ligament-like tissue, positive to Col XII, was observed in ACC transplants, which was absent in DPSC transplants. CONCLUSIONS ACCs and DPSCs showed similar stemness, proliferation rate, and hard tissue-forming capacity. The notable difference was the periodontal ligament-like fiber-forming capacity of ACCs, which indicates the presence of various lineages of stem cells in the apical complex compared with the coronal pulp. Regarding regeneration of the dentin-pulp complex, the coronal pulp can be a suitable source of stem cells considering its homogenous lineages of cells and favorable osteo/odontogenic differentiation potential.
Collapse
Affiliation(s)
- Min-Kyoung Park
- Department of Pediatric Dentistry, College of Dentistry, Yonsei University, Seoul, Republic of Korea
| | - Seunghye Kim
- Department of Pediatric Dentistry, Institute of Oral Health Science, Ajou University, School of Medicine, Suwon, Republic of Korea
| | - Mijeong Jeon
- Oral Science Research Center, College of Dentistry, Yonsei University, Seoul, Republic of Korea
| | - Ui-Won Jung
- Department of Periodontology, Research Institute for Periodontal Regeneration, College of Dentistry, Yonsei University, Seoul, Republic of Korea
| | - Jae-Ho Lee
- Department of Pediatric Dentistry, College of Dentistry, Yonsei University, Seoul, Republic of Korea
| | - Hyung-Jun Choi
- Department of Pediatric Dentistry, College of Dentistry, Yonsei University, Seoul, Republic of Korea
| | - Ja-Eun Choi
- Department of Pediatric Dentistry, College of Dentistry, Yonsei University, Seoul, Republic of Korea
| | - Je Seon Song
- Department of Pediatric Dentistry, College of Dentistry, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
178
|
Characterization of progenitor/stem cell population from human dental socket and their multidifferentiation potential. Cell Tissue Bank 2019; 21:31-46. [PMID: 31807957 DOI: 10.1007/s10561-019-09794-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 11/23/2019] [Indexed: 02/06/2023]
Abstract
Dental stem cells have many applications in medicine, dentistry and stem cell biology in general due to their easy accessibility and low morbidity. A common surgical manoeuvre after a tooth extraction is the dental socket curettage which is necessary to clean the alveolus and favour alveolar bone healing. This procedure can cause very low morbidity compared to bone marrow collection procedures and the collected material is normally discarded. In order to investigate if the tissue obtained by dental socket curettage after a tooth extraction was a feasible alternative source to isolate human stem cells, we isolated and characterized two different stem cell populations based on STRO-1 and CD146 expression. We were able to collect and grow cells from dental socket of vital and non-vital teeth. Both populations were proliferative, clonogenic and expressed STRO-1, CD146, CD90, NG2, PDGFR-β, which are markers found in stem cells, presented in vitro multiline-differentiation into osteogenic, chondrogenic, and adipogenic tissue, and in vivo transplanted cells formed mineralized tissue. Interestingly, STRO-1+ clonogenic cells presented better multidifferentiation than CD146+ cells. Our results showed that mesenchymal stem cells can be isolated from the tiny tissue collected by dental socket curettage after vital and non-vital tooth extraction and suggest that STRO-1 is an important marker to be used to sort cells with multidifferentiation capacity.
Collapse
|
179
|
Mesenchymal stem cell-based bone tissue engineering for veterinary practice. Heliyon 2019; 5:e02808. [PMID: 31844733 PMCID: PMC6895744 DOI: 10.1016/j.heliyon.2019.e02808] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 10/29/2019] [Accepted: 11/07/2019] [Indexed: 01/17/2023] Open
Abstract
Bone tissue engineering has been widely studied and proposed as a promising platform for correcting the bone defects. The applications of mesenchymal stem cell (MSC)-based bone tissue engineering have been investigated in various in vitro and in vivo models. In this regard, the promising animal bone defect models have been employed for illustrating the bone regenerative capacity of MSC-based bone tissue engineering. However, most studies aimed for clinical applications in human. These evidences suggest a knowledge gap to fulfill the accomplishment for veterinary implementation. In this review, the fundamental concept, knowledge, and technology of MSC-based bone tissue engineering focusing on veterinary applications are summarized. In addition, the potential canine MSCs resources for veterinary bone tissue engineering are reviewed, including canine bone marrow-derived MSCs, canine adipose-derived MSCs, and canine dental tissue-derived MSCs. This review will provide a basic and current information for studies aiming for the utilization of MSC-based bone tissue engineering in veterinary practice.
Collapse
|
180
|
Wang Y, Xu J, Meyers CA, Gao Y, Tian Y, Broderick K, Peault B, James AW. PDGFRα marks distinct perivascular populations with different osteogenic potential within adipose tissue. Stem Cells 2019; 38:276-290. [PMID: 31742801 DOI: 10.1002/stem.3108] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/11/2019] [Accepted: 10/09/2019] [Indexed: 12/14/2022]
Abstract
The perivascular niche within adipose tissue is known to house multipotent cells, including osteoblast precursors. However, the identity of perivascular subpopulations that may mineralize or ossify most readily is not known. Here, we utilize inducible PDGFRα (platelet-derived growth factor alpha) reporter animals to identify subpopulations of perivascular progenitor cells. Results showed that PDGFRα-expressing cells are present in four histologic niches within inguinal fat, including two perivascular locations. PDGFRα+ cells are most frequent within the tunica adventitia of arteries and veins, where PDGFRα+ cells populate the inner aspects of the adventitial layer. Although both PDGFRα+ and PDGFRα- fractions are multipotent progenitor cells, adipose tissue-derived PDGFRα+ stromal cells proliferate faster and mineralize to a greater degree than their PDGFRα- counterparts. Likewise, PDGFRα+ ectopic implants reconstitute the perivascular niche and ossify to a greater degree than PDGFRα- cell fractions. Adventicytes can be further grouped into three distinct groups based on expression of PDGFRα and/or CD34. When further partitioned, adventicytes co-expressing PDGFRα and CD34 represented a cell fraction with the highest mineralization potential. Long-term tracing studies showed that PDGFRα-expressing adventicytes give rise to adipocytes, but not to other cells within the vessel wall under homeostatic conditions. However, upon bone morphogenetic protein 2 (BMP2)-induced ossicle formation, descendants of PDGFRα+ cells gave rise to osteoblasts, adipocytes, and "pericyte-like" cells within the ossicle. In sum, PDGFRα marks distinct perivascular osteoprogenitor cell subpopulations within adipose tissue. The identification of perivascular osteoprogenitors may contribute to our improved understanding of pathologic mineralization/ossification.
Collapse
Affiliation(s)
- Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Jiajia Xu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Carolyn A Meyers
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Yongxing Gao
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Ye Tian
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Kristen Broderick
- Department of Plastic Surgery, Johns Hopkins University, Baltimore, Maryland
| | - Bruno Peault
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California.,Center for Cardiovascular Science and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland.,UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California
| |
Collapse
|
181
|
Zhang S, Hu B, Liu W, Wang P, Lv X, Chen S, Liu H, Shao Z. Articular cartilage regeneration: The role of endogenous mesenchymal stem/progenitor cell recruitment and migration. Semin Arthritis Rheum 2019; 50:198-208. [PMID: 31767195 DOI: 10.1016/j.semarthrit.2019.11.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 10/04/2019] [Accepted: 11/01/2019] [Indexed: 01/07/2023]
Abstract
BACKGROUND Trauma- or osteoarthritis-related cartilage damage resulted in functional decline of joints and heavy burden of public health. Recently, the reparative role of mesenchymal stem/progenitor cells (MSCs) in articular cartilage (AC) reconstruction is drawing more and more attention. OBJECTIVE To provide a review on (1) the locations and categories of joint-resident MSCs, (2) the regulation of chondrogenic capacities of MSCs, (3) the migratory approaches of MSCs to diseased AC and regulatory mechanisms. METHODS PubMed and Web of Science were searched for English-language articles related to MSC recruitment and migration for AC repair until June 2019. The presence of various MSCs in or around joints, the potential approaches to diseased AC` and the regenerative capacities of MSCs were reviewed. RESULTS Various intra- and peri-articular MSCs, with inherent migratory potentials, are present in multiple stem cell niches in or around joints. The recruitment and migration of joint-resident MSCs play crucial roles in endogenous AC repair. Multiple recruiting signals, such as chemokines, growth factors, etc., emerge during the development of AC diseases and participate in the regulation of MSC mobilization. Motivated MSCs could migrate into cartilage lesions and then exert multiple reparative potentials, including extracellular matrix (ECM) reconstruction and microenvironment modulation. CONCLUSION In general, AC repair based on endogenous MSC recruitment and migration is a feasible strategy, and a promising research field. Furthermore, endogenous AC repair mediated by native MSCs would provide new opportunities to efficient preventative or therapeutic options for AC diseases.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| | - Binwu Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| | - Weijian Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| | - Peng Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| | - Xiao Lv
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| | - Songfeng Chen
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China.
| | - Hongjian Liu
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China.
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| |
Collapse
|
182
|
Gilbert W, Bragg R, Elmansi AM, McGee-Lawrence ME, Isales CM, Hamrick MW, Hill WD, Fulzele S. Stromal cell-derived factor-1 (CXCL12) and its role in bone and muscle biology. Cytokine 2019; 123:154783. [PMID: 31336263 PMCID: PMC6948927 DOI: 10.1016/j.cyto.2019.154783] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/08/2019] [Accepted: 07/16/2019] [Indexed: 02/07/2023]
Abstract
Musculoskeletal disorders are the leading cause of disability worldwide; two of the most prevalent of which are osteoporosis and sarcopenia. Each affect millions in the aging population across the world and the associated morbidity and mortality contributes to billions of dollars in annual healthcare cost. Thus, it is important to better understand the underlying pathologic mechanisms of the disease process. Regulatory chemokine, CXCL12, and its receptor, CXCR4, are recognized to be essential in the recruitment, localization, maintenance, development and differentiation of progenitor stem cells of the musculoskeletal system. CXCL12 signaling results in the development and functional ability of osteoblasts, osteoclasts, satellite cells and myoblasts critical to maintaining musculoskeletal homeostasis. Interestingly, one suggested pathologic mechanism of osteoporosis and sarcopenia is a decline in the regenerative capacity of musculoskeletal progenitor stem cells. Thus, because CXCL12 is critical to progenitor function, a disruption in the CXCL12 signaling axis might play a distinct role in these pathological processes. Therefore, in this article, we perform a review of CXCL12, its physiologic and pathologic function in bone and muscle, and potential targets for therapeutic development.
Collapse
Affiliation(s)
- William Gilbert
- Department of Orthopaedic Surgery, Augusta University, Augusta, GA 30912, United States
| | - Robert Bragg
- Department of Orthopaedic Surgery, Augusta University, Augusta, GA 30912, United States
| | - Ahmed M Elmansi
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States
| | - Meghan E McGee-Lawrence
- Department of Orthopaedic Surgery, Augusta University, Augusta, GA 30912, United States; Cell Biology and Anatomy, Augusta University, Augusta, GA 30912, United States
| | - Carlos M Isales
- Department of Orthopaedic Surgery, Augusta University, Augusta, GA 30912, United States; Department of Medicine, Augusta University, Augusta, GA 30912, United States
| | - Mark W Hamrick
- Department of Orthopaedic Surgery, Augusta University, Augusta, GA 30912, United States; Cell Biology and Anatomy, Augusta University, Augusta, GA 30912, United States
| | - William D Hill
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States; Ralph H Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States
| | - Sadanand Fulzele
- Department of Orthopaedic Surgery, Augusta University, Augusta, GA 30912, United States; Cell Biology and Anatomy, Augusta University, Augusta, GA 30912, United States.
| |
Collapse
|
183
|
Rajan S, Ljunggren A, Manton DJ, Björkner AE, McCullough M. Post-mitotic odontoblasts in health, disease, and regeneration. Arch Oral Biol 2019; 109:104591. [PMID: 31710968 DOI: 10.1016/j.archoralbio.2019.104591] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 10/09/2019] [Accepted: 10/20/2019] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Description of the odontoblast lifecycle, an overview of the known complex molecular interactions that occur when the health of the dental pulp is challenged and the current and future management strategies on vital and non-vital teeth. METHODS A literature search of the electronic databases included MEDLINE (1966-April 2019), CINAHL (1982-April 2019), EMBASE and EMBASE Classic (1947-April 2019), and hand searches of references retrieved were undertaken using the following MESH terms 'odontoblast*', 'inflammation', 'dental pulp*', 'wound healing' and 'regenerative medicine'. RESULTS Odontoblasts have a sensory and mechano-transduction role so as to detect external stimuli that challenge the dental pulp. On detection, odontoblasts stimulate the innate immunity by activating defence mechanisms key in the healing and repair mechanisms of the tooth. A better understanding of the role of odontoblasts within the dental pulp complex will allow an opportunity for biological management to remove the cause of the insult to the dental pulp, modulate the inflammatory process, and promote the healing and repair capabilities of the tooth. Current strategies include use of conventional dental pulp medicaments while newer methods include bioactive molecules, epigenetic modifications and tissue engineering. CONCLUSION Regenerative medicine methods are in their infancy and experimental stages at best. This review highlights the future direction of dental caries management and consequently research.
Collapse
Affiliation(s)
- S Rajan
- The University of Melbourne, Australia.
| | | | - D J Manton
- The University of Melbourne, Australia; Centrum voor Tandheelkunde en Mondzorgkunde, UMCG, University of Groningen, the Netherlands
| | | | | |
Collapse
|
184
|
Coates DE, Alansary M, Friedlander L, Zanicotti DG, Duncan WJ. Dental pulp stem cells in serum-free medium for regenerative medicine. J R Soc N Z 2019. [DOI: 10.1080/03036758.2019.1673447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Dawn E. Coates
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Mohammad Alansary
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Lara Friedlander
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Diogo G. Zanicotti
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Warwick J. Duncan
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| |
Collapse
|
185
|
Fawzy El-Sayed KM, Elsalawy R, Ibrahim N, Gadalla M, Albargasy H, Zahra N, Mokhtar S, El Nahhas N, El Kaliouby Y, Dörfer CE. The Dental Pulp Stem/Progenitor Cells-Mediated Inflammatory-Regenerative Axis. TISSUE ENGINEERING PART B-REVIEWS 2019; 25:445-460. [DOI: 10.1089/ten.teb.2019.0106] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Karim M. Fawzy El-Sayed
- Oral Medicine and Periodontology Department, Faculty of Oral and Dental Medicine, Cairo University, Cairo, Egypt
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, Kiel, Germany
| | | | | | | | | | - Nehal Zahra
- Faculty of Dentistry, New Giza University, Giza, Egypt
| | | | | | | | - Christof E. Dörfer
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, Kiel, Germany
| |
Collapse
|
186
|
Ambrosi TH, Longaker MT, Chan CKF. A Revised Perspective of Skeletal Stem Cell Biology. Front Cell Dev Biol 2019; 7:189. [PMID: 31572721 PMCID: PMC6753172 DOI: 10.3389/fcell.2019.00189] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 08/23/2019] [Indexed: 12/20/2022] Open
Abstract
Bone-related maladies are a major health burden on modern society. Loss of skeletal integrity and regeneration capacity through aging, obesity, and disease follows from a detrimental shift in bone formation and resorption dynamics. Targeting tissue-resident adult stem cells offers a potentially innovative paradigm in the development of therapeutic strategies against organ dysfunction. While the essential role of skeletal stem cells (SSCs) for development, growth, and maintenance of the skeleton has been generally established, a common consensus on the exact identity and definition of a pure bona fide SSC population remains elusive. The controversies stem from conflicting results between different approaches and criteria for isolation, detection, and functional evaluation; along with the interchangeable usage of the terms SSC and "mesenchymal stromal/stem cell (MSC)". A great number of prospective bone-forming stem cell populations have been reported with various characteristic markers, often describing overlapping cell populations with widely unexplored heterogeneity, species specificity, and distribution at distinct skeletal sites, bone regions, and microenvironments, thereby creating confusion that may complicate future advances in the field. In this review, we examine the state-of-the-art knowledge of SSC biology and try to establish a common ground for the definition and terminology of specific bone-resident stem cells. We also discuss recent advances in the identification of highly purified SSCs, which will allow detailed interrogation of SSC diversity and regulation at the single-cell level.
Collapse
Affiliation(s)
- Thomas H Ambrosi
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Michael T Longaker
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States.,Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Charles K F Chan
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States.,Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
187
|
Naji A, Eitoku M, Favier B, Deschaseaux F, Rouas-Freiss N, Suganuma N. Biological functions of mesenchymal stem cells and clinical implications. Cell Mol Life Sci 2019; 76:3323-3348. [PMID: 31055643 PMCID: PMC11105258 DOI: 10.1007/s00018-019-03125-1] [Citation(s) in RCA: 353] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 04/19/2019] [Accepted: 04/30/2019] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs) are isolated from multiple biological tissues-adult bone marrow and adipose tissues and neonatal tissues such as umbilical cord and placenta. In vitro, MSCs show biological features of extensive proliferation ability and multipotency. Moreover, MSCs have trophic, homing/migration and immunosuppression functions that have been demonstrated both in vitro and in vivo. A number of clinical trials are using MSCs for therapeutic interventions in severe degenerative and/or inflammatory diseases, including Crohn's disease and graft-versus-host disease, alone or in combination with other drugs. MSCs are promising for therapeutic applications given the ease in obtaining them, their genetic stability, their poor immunogenicity and their curative properties for tissue repair and immunomodulation. The success of MSC therapy in degenerative and/or inflammatory diseases might depend on the robustness of the biological functions of MSCs, which should be linked to their therapeutic potency. Here, we outline the fundamental and advanced concepts of MSC biological features and underline the biological functions of MSCs in their basic and translational aspects in therapy for degenerative and/or inflammatory diseases.
Collapse
Affiliation(s)
- Abderrahim Naji
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan.
| | - Masamitsu Eitoku
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| | - Benoit Favier
- CEA, DRF-IBFJ, IDMIT, INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, Paris-Sud University, Fontenay-aux-Roses, France
| | - Frédéric Deschaseaux
- STROMALab, Etablissement Français du Sang Occitanie, UMR 5273 CNRS, INSERM U1031, Université de Toulouse, Toulouse, France
| | - Nathalie Rouas-Freiss
- CEA, DRF-Francois Jacob Institute, Research Division in Hematology and Immunology (SRHI), Saint-Louis Hospital, IRSL, UMRS 976, Paris, France
| | - Narufumi Suganuma
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| |
Collapse
|
188
|
Stem cell-based bone and dental regeneration: a view of microenvironmental modulation. Int J Oral Sci 2019; 11:23. [PMID: 31423011 PMCID: PMC6802669 DOI: 10.1038/s41368-019-0060-3] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/28/2019] [Accepted: 06/12/2019] [Indexed: 02/06/2023] Open
Abstract
In modern medicine, bone and dental loss and defects are common and widespread morbidities, for which regenerative therapy has shown great promise. Mesenchymal stem cells, obtained from various sources and playing an essential role in organ development and postnatal repair, have exhibited enormous potential for regenerating bone and dental tissue. Currently, mesenchymal stem cells (MSCs)-based bone and dental regeneration mainly includes two strategies: the rescue or mobilization of endogenous MSCs and the application of exogenous MSCs in cytotherapy or tissue engineering. Nevertheless, the efficacy of MSC-based regeneration is not always fulfilled, especially in diseased microenvironments. Specifically, the diseased microenvironment not only impairs the regenerative potential of resident MSCs but also controls the therapeutic efficacy of exogenous MSCs, both as donors and recipients. Accordingly, approaches targeting a diseased microenvironment have been established, including improving the diseased niche to restore endogenous MSCs, enhancing MSC resistance to a diseased microenvironment and renormalizing the microenvironment to guarantee MSC-mediated therapies. Moreover, the application of extracellular vesicles (EVs) as cell-free therapy has emerged as a promising therapeutic strategy. In this review, we summarize current knowledge regarding the tactics of MSC-based bone and dental regeneration and the decisive role of the microenvironment, emphasizing the therapeutic potential of microenvironment-targeting strategies in bone and dental regenerative medicine.
Collapse
|
189
|
Hsu CY, Salazar MG, Miller S, Meyers C, Ding C, Hardy W, Péault B, James AW. Comparison of Human Tissue Microarray to Human Pericyte Transcriptome Yields Novel Perivascular Cell Markers. Stem Cells Dev 2019; 28:1214-1223. [PMID: 31264500 DOI: 10.1089/scd.2019.0106] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Human perivascular progenitor cells, including pericytes, are well-described multipotent mesenchymal cells giving rise to mesenchymal stem cells in culture. Despite the unique location of pericytes, specific antigens to distinguish human pericytes from other cell types are few. Here, we employed a human tissue microarray (Human Protein Atlas) to identify proteins that are strongly and specifically expressed in a pericytic location within human adipose tissue. Next, these results were cross-referenced with RNA sequencing data from human adipose tissue pericytes, as defined as a fluorescence activated cell sorting (FACS) purified CD146+CD34-CD31-CD45- cell population. Results showed that from 105,532 core biopsies of soft tissue, 229 proteins showed strong and specific perivascular immunoreactivity, the majority of which (155) were present in the tunica intima. Next, cross-referencing with the transcriptome of FACS-derived CD146+ pericytes yielded 25 consistently expressed genes/proteins, including 18 novel antigens. A majority of these transcripts showed maintained expression after culture propagation (56% of genes). Interestingly, many novel antigens within pericytes are regulators of osteogenic differentiation. In sum, our study demonstrates the existence of novel pericyte markers, some of which are conserved in culture that may be useful for future efforts to typify, isolate, and characterize human pericytes.
Collapse
Affiliation(s)
- Ching Yun Hsu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Mario Gomez Salazar
- Center for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom.,MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Sarah Miller
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Carolyn Meyers
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Catherine Ding
- Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Winters Hardy
- Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Bruno Péault
- Center for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom.,MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom.,Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland.,Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
190
|
Mesenchymal Stem Cells in Homeostasis and Systemic Diseases: Hypothesis, Evidences, and Therapeutic Opportunities. Int J Mol Sci 2019; 20:ijms20153738. [PMID: 31370159 PMCID: PMC6696100 DOI: 10.3390/ijms20153738] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 07/29/2019] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are present in all organs and tissues, playing a well-known function in tissue regeneration. However, there is also evidence indicating a broader role of MSCs in tissue homeostasis. In vivo studies have shown MSC paracrine mechanisms displaying proliferative, immunoregulatory, anti-oxidative, or angiogenic activity. In addition, recent studies also demonstrate that depletion and/or dysfunction of MSCs are associated with several systemic diseases, such as lupus, diabetes, psoriasis, and rheumatoid arthritis, as well as with aging and frailty syndrome. In this review, we hypothesize about the role of MSCs as keepers of tissue homeostasis as well as modulators in a variety of inflammatory and degenerative systemic diseases. This scenario opens the possibility for the use of secretome-derived products from MSCs as new therapeutic agents in order to restore tissue homeostasis, instead of the classical paradigm "one disease, one drug".
Collapse
|
191
|
Sun X, Meng L, Qiao W, Yang R, Gao Q, Peng Y, Bian Z. Vascular endothelial growth factor A/Vascular endothelial growth factor receptor 2 axis promotes human dental pulp stem cell migration via the FAK/PI3K/Akt and p38 MAPK signalling pathways. Int Endod J 2019; 52:1691-1703. [PMID: 31267530 DOI: 10.1111/iej.13179] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 06/27/2019] [Indexed: 01/04/2023]
Abstract
AIM To investigate the effects of vascular endothelial growth factor A (VEGFA) and the underlying molecular mechanisms on the migration of human dental pulp stem cells (hDPSCs). METHODOLOGY The expression of VEGFA in inflammatory pulp tissue and lipopolysaccharide (LPS)-stimulated dental pulp cells was examined by immunofluorescence staining and qRT-PCR. The migration of hDPSCs was detected using transwell migration and wound healing assays. The activation of FAK, PI3K, Akt and p38 signalling was evaluated by Western blot analysis. Silence RNA (siRNA) technology was utilized to knockdown the expression of VEGFR1 (Flt-1) and VEGFR2 (Flk-1/KDR). PF573228 (inhibitor of FAK), LY294002 (inhibitor of PI3K), SB203580 (inhibitor of p38) and SU5416 (inhibitor of VEGFR2) were employed to investigate the effect of VEGFA on the migratory mechanism of hDPSCs. Data were analysed statistically using the Student's t-test or one-way ANOVA. RESULTS The expression levels of VEGFA in inflammatory pulp tissue in vivo and LPS-stimulated dental pulp cells in vitro were significantly greater than those in the control groups (P < 0.05). Vascular endothelial growth factor A promoted the migration of hDPSCs in a concentration-dependent manner. Several signalling pathways, including FAK, PI3K, Akt and p38, were activated by VEGFA in a dose- and time-dependent manner in hDPSCs. The VEGFA-induced migration of hDPSCs was significantly inhibited with drug inhibitors such as PF573228, LY294002, SB203580 or SU5416 (P < 0.05). These signalling pathways activated by VEGFA stimulation were significantly suppressed by pre-treatment with inhibitor of VEGFR2 (SU5416) or transfection with siRNA of VRGFR2 (P < 0.05) but not VEGFR1 siRNA. CONCLUSIONS Vascular endothelial growth factor A/VEGFR2 axis promoted the migration of hDPSCs via the FAK/PI3K/Akt and p38 MAPK signalling pathways. These findings reveal a novel molecular mechanism for cell migration of hDPSCs, which may contribute to the remodelling of pulp tissue and dentine.
Collapse
Affiliation(s)
- X Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - L Meng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - W Qiao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - R Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Q Gao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Y Peng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Z Bian
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
192
|
Xiao J, Cao P, Wang C, Huang D, Lian M, Song Y, Yin W, Zheng K, Gu Z, Gu Y, Feng G, Feng X. The Forkhead Box C1, a Novel Negative Regulator of Osteogenesis, Plays a Crucial Role in Odontogenic Differentiation of Dental Pulp Stem Cells. Cell Reprogram 2019; 20:312-319. [PMID: 30277823 DOI: 10.1089/cell.2018.0011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The forkhead box C1 (Foxc1) protein, a member of the forkhead/winged helix transcription factor family, is required in stem cell developmental processes. Recently, multiple studies have indicated the crucial role of Foxc1 in mesenchymal stem cell differentiation, but the precise effects and mechanisms on dental pulp stem cells (DPSCs) remain unclear. In this study, we evaluate the role of Foxc1 on the odontogenic differentiation and proliferation of DPSCs. Our results show that Foxc1 decreases time dependently in odontogenic differentiation of DPSCs. Meanwhile, overexpression of Foxc1 could significantly inhibit the mineralization of DPSCs and the expression of odontogenic-related genes, such as runt-related transcription factor 2 (Runx2), dentin sialophosphoprote (DSPP), and dentin matrix acidic phosphoprotein 1 (DMP-1). Foxc1 overexpression does not significantly alter the proliferation of DPSCs. In addition, Foxc1 reduces the expression of p-Smad1/5, an important modulator of bone morphogenetic protein (BMP)/Smad signaling pathway, inhibiting BMP/Smad signaling pathway. In conclusion, our data demonstrated that Foxc1 inhibits odontogenic differentiation of DPSCs and odontogenic-related gene expression through the BMP/Smad signaling pathway which may be useful for the dental regeneration and repair.
Collapse
Affiliation(s)
- Jingwen Xiao
- 1 Department of Stomatology, Affiliated Hospital of Nantong University, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, Jiangsu, China
| | - Peipei Cao
- 1 Department of Stomatology, Affiliated Hospital of Nantong University, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, Jiangsu, China
| | - Chenfei Wang
- 1 Department of Stomatology, Affiliated Hospital of Nantong University, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, Jiangsu, China
| | - Dan Huang
- 1 Department of Stomatology, Affiliated Hospital of Nantong University, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, Jiangsu, China
| | - Min Lian
- 1 Department of Stomatology, Affiliated Hospital of Nantong University, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, Jiangsu, China
| | - Yihua Song
- 1 Department of Stomatology, Affiliated Hospital of Nantong University, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, Jiangsu, China
| | - Weiwei Yin
- 2 Department of Stomatology, Stomatological Hospital of Nantong City , Nantong, Jiangsu, China
| | - Ke Zheng
- 1 Department of Stomatology, Affiliated Hospital of Nantong University, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, Jiangsu, China
| | - Zhifeng Gu
- 3 Department of Rheumatology, Affiliated Hospital of Nantong University , Nantong, Jiangsu, China
| | - Yongchun Gu
- 4 Department of Stomatology, First People's Hospital of Wujiang District, Nantong University , Suzhou, Jiangsu, China
| | - Guijuan Feng
- 1 Department of Stomatology, Affiliated Hospital of Nantong University, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, Jiangsu, China
| | - Xingmei Feng
- 1 Department of Stomatology, Affiliated Hospital of Nantong University, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, Jiangsu, China
| |
Collapse
|
193
|
Abstract
Cells have been identified in postnatal tissues that, when isolated from multiple mesenchymal compartments, can be stimulated in vitro to give rise to cells that resemble mature mesenchymal phenotypes, such as odontoblasts, osteoblasts, adipocytes, and myoblasts. This has made these adult cells, collectively called mesenchymal stem cells (MSCs), strong candidates for fields such as tissue engineering and regenerative medicine. Based on evidence from in vivo genetic lineage-tracing studies, pericytes have been identified as a source of MSC precursors in vivo in multiple organs, in response to injury or during homeostasis. Questions of intense debate and interest in the field of tissue engineering and regenerative studies include the following: 1) Are all pericytes, irrespective of tissue of isolation, equal in their differentiation potential? 2) What are the mechanisms that regulate the differentiation of MSCs? To gain a better understanding of the latter, recent work has utilized ChIP-seq (chromatin immunoprecipitation followed by sequencing) to reconstruct histone landscapes. This indicated that for dental pulp pericytes, the odontoblast-specific gene Dspp was found in a transcriptionally permissive state, while in bone marrow pericytes, the osteoblast-specific gene Runx2 was primed for expression. RNA sequencing has also been utilized to further characterize the 2 pericyte populations, and results highlighted that dental pulp pericytes are already precommitted to an odontoblast fate based on enrichment analysis indicating overrepresentation of key odontogenic genes. Furthermore, ChIP-seq analysis of the polycomb repressive complex 1 component RING1B indicated that this complex is likely to be involved in inhibiting inappropriate differentiation, as it localized to a number of loci of key transcription factors that are needed for the induction of adipogenesis, chondrogenesis, or myogenesis. In this review, we highlight recent data elucidating molecular mechanisms that indicate that pericytes can be tissue-specific precommitted MSC precursors in vivo and that this precommitment is a major driving force behind MSC differentiation.
Collapse
Affiliation(s)
- V Yianni
- 1 Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | - P T Sharpe
- 1 Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| |
Collapse
|
194
|
De Luca M, Aiuti A, Cossu G, Parmar M, Pellegrini G, Robey PG. Advances in stem cell research and therapeutic development. Nat Cell Biol 2019; 21:801-811. [PMID: 31209293 DOI: 10.1038/s41556-019-0344-z] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 05/09/2019] [Indexed: 12/12/2022]
Abstract
Despite many reports of putative stem-cell-based treatments in genetic and degenerative disorders or severe injuries, the number of proven stem cell therapies has remained small. In this Review, we survey advances in stem cell research and describe the cell types that are currently being used in the clinic or are close to clinical trials. Finally, we analyse the scientific rationale, experimental approaches, caveats and results underpinning the clinical use of such stem cells.
Collapse
Affiliation(s)
- Michele De Luca
- Center for Regenerative Medicine "Stefano Ferrari", Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget) and Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Giulio Cossu
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Malin Parmar
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Graziella Pellegrini
- Center for Regenerative Medicine "Stefano Ferrari", Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Pamela Gehron Robey
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| |
Collapse
|
195
|
Photobiomodulatory effect delivered by low-level laser on dental pulp stem cell differentiation for osteogenic lineage. ACTA ACUST UNITED AC 2019. [DOI: 10.1007/s41547-019-00066-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
196
|
New Insights on Properties and Spatial Distributions of Skeletal Stem Cells. Stem Cells Int 2019; 2019:9026729. [PMID: 31281389 PMCID: PMC6589297 DOI: 10.1155/2019/9026729] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 05/13/2019] [Indexed: 02/06/2023] Open
Abstract
Skeletal stem cells (SSCs) are postnatal self-renewing, multipotent, and skeletal lineage-committed progenitors that are capable of giving rise to cartilage, bone, and bone marrow stroma including marrow adipocytes and stromal cells in vitro and in an exogenous environment after transplantation in vivo. Identifying and isolating defined SSCs as well as illuminating their spatiotemporal properties contribute to our understating of skeletal biology and pathology. In this review, we revisit skeletal stem cells identified most recently and systematically discuss their origin and distributions.
Collapse
|
197
|
Qiryaqoz Z, Timilsina S, Czarnowski D, Krebsbach PH, Villa‐Diaz LG. Identification of biomarkers indicative of functional skeletal stem cells. Orthod Craniofac Res 2019; 22 Suppl 1:192-198. [DOI: 10.1111/ocr.12260] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 12/04/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Zeena Qiryaqoz
- Department of Biological SciencesOakland University Rochester Michigan
| | - Suraj Timilsina
- Department of Biological SciencesOakland University Rochester Michigan
| | - Daniel Czarnowski
- Department of Biological SciencesOakland University Rochester Michigan
| | - Paul H. Krebsbach
- School of DentistryUniversity of California, Los Angeles Los Angeles California
| | | |
Collapse
|
198
|
Lin H, Sohn J, Shen H, Langhans MT, Tuan RS. Bone marrow mesenchymal stem cells: Aging and tissue engineering applications to enhance bone healing. Biomaterials 2019; 203:96-110. [PMID: 29980291 PMCID: PMC6733253 DOI: 10.1016/j.biomaterials.2018.06.026] [Citation(s) in RCA: 256] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/18/2018] [Accepted: 06/19/2018] [Indexed: 12/11/2022]
Abstract
Bone has well documented natural healing capacity that normally is sufficient to repair fractures and other common injuries. However, the properties of bone change throughout life, and aging is accompanied by increased incidence of bone diseases and compromised fracture healing capacity, which necessitate effective therapies capable of enhancing bone regeneration. The therapeutic potential of adult mesenchymal stem cells (MSCs) for bone repair has been long proposed and examined. Actions of MSCs may include direct differentiation to become bone cells, attraction and recruitment of other cells, or creation of a regenerative environment via production of trophic growth factors. With systemic aging, MSCs also undergo functional decline, which has been well investigated in a number of recent studies. In this review, we first describe the changes in MSCs during aging and discuss how these alterations can affect bone regeneration. We next review current research findings on bone tissue engineering, which is considered a promising and viable therapeutic solution for structural and functional restoration of bone. In particular, the importance of MSCs and bioscaffolds is highlighted. Finally, potential approaches for the prevention of MSC aging and the rejuvenation of aged MSC are discussed.
Collapse
Affiliation(s)
- Hang Lin
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jihee Sohn
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - He Shen
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Division of Nanobiomedicine, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, China
| | - Mark T Langhans
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
199
|
Gancheva MR, Kremer KL, Gronthos S, Koblar SA. Using Dental Pulp Stem Cells for Stroke Therapy. Front Neurol 2019; 10:422. [PMID: 31110489 PMCID: PMC6501465 DOI: 10.3389/fneur.2019.00422] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 04/08/2019] [Indexed: 12/26/2022] Open
Abstract
Stroke is a leading cause of permanent disability world-wide, but aside from rehabilitation, there is currently no clinically-proven pharmaceutical or biological agent to improve neurological disability. Cell-based therapies using stem cells, such as dental pulp stem cells, are a promising alternative for treatment of neurological diseases, including stroke. The ischaemic environment in stroke affects multiple cell populations, thus stem cells, which act through cellular and molecular mechanisms, are promising candidates. The most common stem cell population studied in the neurological setting has been mesenchymal stem cells due to their accessibility. However, it is believed that neural stem cells, the resident stem cell of the adult brain, would be most appropriate for brain repair. Using reprogramming strategies, alternative sources of neural stem and progenitor cells have been explored. We postulate that a cell of closer origin to the neural lineage would be a promising candidate for reprogramming and modification towards a neural stem or progenitor cell. One such candidate population is dental pulp stem cells, which reside in the root canal of teeth. This review will focus on the neural potential of dental pulp stem cells and their investigations in the stroke setting to date, and include an overview on the use of different sources of neural stem cells in preclinical studies and clinical trials of stroke.
Collapse
Affiliation(s)
- Maria R. Gancheva
- Stroke Research Programme Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Karlea L. Kremer
- Stroke Research Programme Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Simon A. Koblar
- Stroke Research Programme Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Central Adelaide Local Health Network, Adelaide, SA, Australia
| |
Collapse
|
200
|
Ramazzotti G, Ratti S, Fiume R, Follo MY, Billi AM, Rusciano I, Owusu Obeng E, Manzoli L, Cocco L, Faenza I. Phosphoinositide 3 Kinase Signaling in Human Stem Cells from Reprogramming to Differentiation: A Tale in Cytoplasmic and Nuclear Compartments. Int J Mol Sci 2019; 20:ijms20082026. [PMID: 31022972 PMCID: PMC6514809 DOI: 10.3390/ijms20082026] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 04/19/2019] [Accepted: 04/21/2019] [Indexed: 12/11/2022] Open
Abstract
Stem cells are undifferentiated cells that can give rise to several different cell types and can self-renew. Given their ability to differentiate into different lineages, stem cells retain huge therapeutic potential for regenerative medicine. Therefore, the understanding of the signaling pathways involved in stem cell pluripotency maintenance and differentiation has a paramount importance in order to understand these biological processes and to develop therapeutic strategies. In this review, we focus on phosphoinositide 3 kinase (PI3K) since its signaling pathway regulates many cellular processes, such as cell growth, proliferation, survival, and cellular transformation. Precisely, in human stem cells, the PI3K cascade is involved in different processes from pluripotency and induced pluripotent stem cell (iPSC) reprogramming to mesenchymal and oral mesenchymal differentiation, through different and interconnected mechanisms.
Collapse
Affiliation(s)
- Giulia Ramazzotti
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Stefano Ratti
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Roberta Fiume
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Matilde Yung Follo
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Anna Maria Billi
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Isabella Rusciano
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Eric Owusu Obeng
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Lucia Manzoli
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Lucio Cocco
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| | - Irene Faenza
- Department of Biomedical Sciences, University of Bologna, Via Irnerio, 48, 40126 Bologna, Italy.
| |
Collapse
|