1
|
Dupuy S, Salvador J, Morille M, Noël D, Belamie E. Control and interplay of scaffold-biomolecule interactions applied to cartilage tissue engineering. Biomater Sci 2025; 13:1871-1900. [PMID: 40052975 DOI: 10.1039/d5bm00049a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Cartilage tissue engineering based on the combination of biomaterials, adult or stem cells and bioactive factors is a challenging approach for regenerative medicine with the aim of achieving the formation of a functional neotissue stable in the long term. Various 3D scaffolds have been developed to mimic the extracellular matrix environment and promote cartilage repair. In addition, bioactive factors have been extensively employed to induce and maintain the cartilage phenotype. However, the spatiotemporal control of bioactive factor release remains critical for maximizing the regenerative potential of multipotent cells, such as mesenchymal stromal cells (MSCs), and achieving efficient chondrogenesis and sustained tissue homeostasis, which are essential for the repair of hyaline cartilage. Despite advances, the effective delivery of bioactive factors is limited by challenges such as insufficient retention at the site of injury and the loss of therapeutic efficacy due to uncontrolled drug release. These limitations have prompted research on biomolecule-scaffold interactions to develop advanced delivery systems that provide sustained release and controlled bioavailability of biological factors, thereby improving therapeutic outcomes. This review focuses specifically on biomaterials (natural, hybrid and synthetic) and biomolecules (molecules, proteins, nucleic acids) of interest for cartilage engineering. Herein, we review in detail the approaches developed to maintain the biomolecules in scaffolds and control their release, based on their chemical nature and structure, through steric, non-covalent and/or covalent interactions, with a view to their application in cartilage repair.
Collapse
Affiliation(s)
- Silouane Dupuy
- ICGM, University of Montpellier, CNRS, ENSCM, Montpellier, France.
- IRMB, University of Montpellier, INSERM, Montpellier, France
| | - Jérémy Salvador
- ICGM, University of Montpellier, CNRS, ENSCM, Montpellier, France.
- EPHE, PSL Research University, 75014 Paris, France
- IRMB, University of Montpellier, INSERM, Montpellier, France
| | - Marie Morille
- ICGM, University of Montpellier, CNRS, ENSCM, Montpellier, France.
| | - Danièle Noël
- IRMB, University of Montpellier, INSERM, Montpellier, France
| | - Emmanuel Belamie
- ICGM, University of Montpellier, CNRS, ENSCM, Montpellier, France.
- EPHE, PSL Research University, 75014 Paris, France
| |
Collapse
|
2
|
Zhu F, Ji L, Dai K, Deng S, Wang J, Liu C. In situ licensing of mesenchymal stem cell immunomodulatory function via BMP-2 induced developmental process. Proc Natl Acad Sci U S A 2024; 121:e2410579121. [PMID: 39565311 PMCID: PMC11621467 DOI: 10.1073/pnas.2410579121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024] Open
Abstract
The immunomodulatory function of mesenchymal stem cells (MSCs) is plastic and susceptible to resident microenvironment in vivo or inflammatory factors in vitro. We propose a unique method to enhance the immunoregulatory functions of mesenchymal stem cells (MSCs) through an artificially controllable in vivo inflammatory microenvironment generated by biomaterials loaded with BMP-2 that induce bone development. MSCs activated through this method effectively induce M1 macrophage polarization toward the M2 phenotype, promote differentiation of naïve T cells into regulatory T cells, and inhibit the proliferation of activated T cells via prostaglandin E2 (PGE2) secretion. This in vivo licensing not only preserves the immunogenicity of MSCs but also alters DNA methylation patterns, enabling MSCs to exhibit immunoregulatory effects with epigenetic memory. Validation in a mouse colitis model demonstrated their therapeutic efficacy and long-term viability. Furthermore, we found that the material composition influences the inflammatory response during development, with polysaccharide-based biomaterials proving advantageous over protein-based materials in establishing an inflammatory niche conducive to MSC activity. These findings underscore the potential of tissue engineering to create in vivo environments that license MSCs, offering a strategic avenue to enhance MSC-based therapies for addressing significant immune disorders.
Collapse
Affiliation(s)
- Fuwei Zhu
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai200237, People’s Republic of China
- School of Materials Science and Engineering, Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Luli Ji
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai200237, People’s Republic of China
- School of Materials Science and Engineering, Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Kai Dai
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai200237, People’s Republic of China
- School of Materials Science and Engineering, Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Shunshu Deng
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai200237, People’s Republic of China
- School of Materials Science and Engineering, Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Jing Wang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai200237, People’s Republic of China
- School of Materials Science and Engineering, Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai200237, People’s Republic of China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Changsheng Liu
- School of Materials Science and Engineering, Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai200237, People’s Republic of China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai200237, People’s Republic of China
- School of Materials Science and Engineering, Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| |
Collapse
|
3
|
Weston JD, Austin B, Levis H, Zitnay J, Weiss JA, Lawrence B, Bowles RD. Toward the Development of a Tissue Engineered Gradient Utilizing CRISPR-Guided Gene Modulation. Tissue Eng Part A 2024; 30:525-535. [PMID: 38323556 DOI: 10.1089/ten.tea.2023.0352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024] Open
Abstract
Cellular, compositional, and mechanical gradients are found throughout biological tissues, especially in transition zones between tissue types. Yet, strategies to engineer such gradients have proven difficult due to the complex nature of these tissues. Current strategies for tissue engineering complex gradients often utilize stem cells; however, these multipotent cells require direction from environmental cues, which can be difficult to control both in vitro and in vivo. In this study, we utilize clustered regularly-interspaced short palindromic repeats (CRISPR)-guided gene modulation to direct the differentiation of multipotent adipose-derived stem cells (ASCs) to demonstrate the effectiveness of CRISPR-engineered cells in tissue engineering applications. Specifically, we screen CRISPR-interference (CRISPRi) constructs targeting the promotors of selected osteogenic inhibitors and demonstrate that ASC osteogenic differentiation and mineral deposition can be regulated with CRISPRi targeting of Noggin without the use of exogenous growth factors in tissue engineered constructs. As a proof of concept, we combine three technologies developed out of our laboratories to demonstrate the controlled deposition of these engineered cells in a gradient with CRISPR-activation multiplex-engineered aggrecan/collagen type-II-chondrogenic ASCs on a high density anisotropic type I collagen construct to create a cell and tissue gradient similar to the fibrocartilage-to-mineralized-fibrocartilage gradient in the enthesis. Our results display the promise of CRISPR-engineered ASCs to produce tissue gradients, similar to what is observed in native tissue.
Collapse
Affiliation(s)
- Jacob D Weston
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Brooke Austin
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Hunter Levis
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Jared Zitnay
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Jeffrey A Weiss
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Brandon Lawrence
- Department of Orthopedic Surgery, University of Utah, Salt Lake City, Utah, USA
| | - Robby D Bowles
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
4
|
Sessler TM, Beier JP, Villwock S, Jonigk D, Dahl E, Ruhl T. Genetic deletion of ITIH5 leads to increased development of adipose tissue in mice. Biol Res 2024; 57:58. [PMID: 39198923 PMCID: PMC11360682 DOI: 10.1186/s40659-024-00530-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 07/18/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Adipocytokines play a pivotal role in maintaining adipose tissue homeostasis by regulating cellular metabolism, proliferation, differentiation, and secretory activity. These soluble factors are relevant components for healthy adipose tissue, while their deficiency is closely associated with the development of obesity and related metabolic diseases, e.g., chronic inflammation. In human adipose tissue, inter-α-trypsin inhibitor heavy chain 5 (ITIH5) is expressed in proportion to the development of adipose tissue, i.e., the individual's BMI. Thus, ITIH5 has been proposed to be an inert marker of human obesity. However, when applied to adipose stem cells in vitro, recombinant (r)ITIH5 protein inhibited proliferation and adipogenesis, suggesting that ITIH5 negatively affects the development of fat mass. We now tested the role of ITIH5 in vivo and compared ITIH5+/+ wildtype with ITIH5-/- knockout mice. RESULTS Genetic deletion of ITIH5 significantly increased adipose tissue mass relative to animal bodyweight (p < 0.05). Next, we characterized adipose stem cells (ASCs) from both genotypes in vitro. ITIH5-/- cells exhibited increased proliferation and adipogenic differentiation (p < 0.001), which could explain the increase in adipose tissue in vivo. Furthermore, ASCs from ITIH5-/- animals were more responsive to stimulation with inflammatory mediators, i.e., these cells released greater amounts of IL-6 and MCP-1 (p < 0.001). Importantly, the application of the rITIH5 protein reversed the observed knockout effects in ASCs. CONCLUSIONS Our data suggest that ITIH5 potently regulates adipose tissue development and homeostasis by modulating ASC biology in mice. In addition, the effect of the rITIH5 protein underscores its potential as a therapeutic agent to correct the adipose tissue dysregulation often associated with obesity and metabolic disorders.
Collapse
Affiliation(s)
- Thomas M Sessler
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Justus P Beier
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Sophia Villwock
- Institute of Pathology, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Danny Jonigk
- Institute of Pathology, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
- Biomedical Research in End-stage and Obstructive Lung Disease Hannover (BREATH) of the German Center for Lung Research (DZL), Hanover, Germany
| | - Edgar Dahl
- Institute of Pathology, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Tim Ruhl
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany.
| |
Collapse
|
5
|
Schumacher A, Mucha P, Puchalska I, Deptuła M, Wardowska A, Tymińska A, Filipowicz N, Mieczkowska A, Sachadyn P, Piotrowski A, Pikuła M, Cichorek M. Angiopoietin-like growth factor-derived peptides as biological activators of adipose-derived mesenchymal stromal cells. Biomed Pharmacother 2024; 177:117052. [PMID: 38943988 DOI: 10.1016/j.biopha.2024.117052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024] Open
Abstract
Adipose-derived mesenchymal stromal cells (AD-MSCs) are an essential issue in modern medicine. Extensive preclinical and clinical studies have shown that mesenchymal stromal/stem cells, including AD-MSCs, have specific properties (ability to differentiate into other cells, recruitment to the site of injury) of particular importance in the regenerative process. Ongoing research aims to elucidate factors supporting AD-MSC culture and differentiation in vitro. Angiopoietin-like proteins (ANGPTLs), known for their pleiotropic effects in lipid and glucose metabolism, may play a significant role in this context. Regeneration is a complex and dynamic process controlled by many factors. ANGPTL6 (Angiopoietin-related growth factor, AGF), among many activities modulated the biological activity of stem cells. This study examined the influence of synthesized AGF-derived peptides, designated as AGF9 and AGF27, on AD-MSCs. AGF9 and AGF27 enhanced the viability and migration of AD-MSCs and acted as a chemotactic factor for these cells. AGF9 stimulated chondrogenesis and lipid synthesis during AD-MSCs differentiation, influenced AD-MSCs cytokine secretion and modulated transcriptome for such basic cell activities as migration, transport of molecules, and apoptosis. The ability of AGF9 to modulate the biological activity of AD-MSCs warrants the consideration of this peptide a noteworthy therapeutic agent that deserves further investigation for applications in regenerative medicine.
Collapse
Affiliation(s)
- Adriana Schumacher
- Division of Embryology, Medical University of Gdansk, Debinki 1 St, Gdansk 80-211, Poland
| | - Piotr Mucha
- Department of Molecular Biochemistry, University of Gdansk, Wita Stwosza 63 St, Gdansk 80-308, Poland
| | - Izabela Puchalska
- Department of Molecular Biochemistry, University of Gdansk, Wita Stwosza 63 St, Gdansk 80-308, Poland
| | - Milena Deptuła
- Division of Embryology, Laboratory of Tissue Engineering and Regenerative Medicine Medical University of Gdansk, Debinki 1 St, Gdansk 80-211, Poland
| | - Anna Wardowska
- Department of Physiopathology, Medical University of Gdansk, Debinki 7 St, Gdansk 80-211, Poland
| | - Agata Tymińska
- Division of Embryology, Medical University of Gdansk, Debinki 1 St, Gdansk 80-211, Poland
| | - Natalia Filipowicz
- International Research Agenda 3P- Medicine Laboratory, Medical University of Gdansk, Debinki 7 St, Gdansk 80-211, Poland
| | - Alina Mieczkowska
- International Research Agenda 3P- Medicine Laboratory, Medical University of Gdansk, Debinki 7 St, Gdansk 80-211, Poland
| | - Paweł Sachadyn
- Laboratory for Regenerative Biotechnology, Gdansk University of Technology, Narutowicza 11/12 St, Gdansk 80-233, Poland
| | - Arkadiusz Piotrowski
- International Research Agenda 3P- Medicine Laboratory, Medical University of Gdansk, Debinki 7 St, Gdansk 80-211, Poland
| | - Michał Pikuła
- Division of Embryology, Laboratory of Tissue Engineering and Regenerative Medicine Medical University of Gdansk, Debinki 1 St, Gdansk 80-211, Poland
| | - Miroslawa Cichorek
- Division of Embryology, Medical University of Gdansk, Debinki 1 St, Gdansk 80-211, Poland.
| |
Collapse
|
6
|
Funke S, Wiggenhauser PS, Grundmeier A, Taha S, Fuchs B, Birt A, Koban K, Giunta RE, Kuhlmann C. Aspirin Stimulates the Osteogenic Differentiation of Human Adipose Tissue-Derived Stem Cells In Vitro. Int J Mol Sci 2024; 25:7690. [PMID: 39062933 PMCID: PMC11277042 DOI: 10.3390/ijms25147690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
This study investigates the impact of acetylsalicylic acid (ASA), also known as aspirin, on adipose tissue-derived stem cells (ASCs), aiming to elucidate its dose-dependent effects on morphology, viability, proliferation, and osteogenic differentiation. Isolated and characterized human ASCs were exposed to 0 µM, 100 µM, 200 µM, 400 µM, 800 µM, 1000 µM, 10,000 µM, and 16,000 µM of ASA in vitro. Cell morphology, viability, and proliferation were evaluated with fluorescent live/dead staining, alamarBlue viability reagent, and CyQUANT® cell proliferation assay, respectively. Osteogenic differentiation under stimulation with 400 µM or 1000 µM of ASA was assessed with alizarin red staining and qPCR of selected osteogenic differentiation markers (RUNX2, SPP1, ALPL, BGLAP) over a 3- and 21-day-period. ASA doses ≤ 1000 µM showed no significant impact on cell viability and proliferation. Live/dead staining revealed a visible reduction in viable cell confluency for ASA concentrations ≥ 1000 µM. Doses of 10,000 µM and 16,000 µM of ASA exhibited a strong cytotoxic and anti-proliferative effect in ASCs. Alizarin red staining revealed enhanced calcium accretion under the influence of ASA, which was macro- and microscopically visible and significant for 1000 µM of ASA (p = 0.0092) in quantification if compared to osteogenic differentiation without ASA addition over a 21-day-period. This enhancement correlated with a more pronounced upregulation of osteogenic markers under ASA exposure (ns). Our results indicate a stimulatory effect of 1000 µM of ASA on the osteogenic differentiation of ASCs. Further research is needed to elucidate the precise molecular mechanisms underlying this effect; however, this discovery suggests promising opportunities for enhancing bone tissue engineering with ASCs as cell source.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Constanze Kuhlmann
- Division of Hand Surgery, Plastic Surgery and Aesthetic Surgery, University Hospital, LMU Munich, Ziemssenstraße 5, 80336 Munich, Germany; (S.F.); (P.S.W.); (A.G.); (S.T.); (B.F.); (A.B.); (K.K.); (R.E.G.)
| |
Collapse
|
7
|
Toyoda M, Fukuda T, Fujimoto R, Kawakami K, Hayashi C, Nakao Y, Watanabe Y, Aoki T, Shida M, Sanui T, Taguchi M, Yamamichi K, Okabe A, Okada T, Oka K, Nakayama K, Nishimura F, Kajioka S. Scaffold-free bone-like 3D structure established through osteogenic differentiation from human gingiva-derived stem cells. Biochem Biophys Rep 2024; 38:101656. [PMID: 38379857 PMCID: PMC10878834 DOI: 10.1016/j.bbrep.2024.101656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/02/2024] [Accepted: 01/27/2024] [Indexed: 02/22/2024] Open
Abstract
Introduction & objectives Stem cell therapy for regenerative medicine has been sincerely investigated, but not still popular although some clinical trials show hopeful results. This therapy is suggested to be a representative candidate such as bone defect due to the accident, iatrogenic resection oncological tumor, congenital disease, and severe periodontitis in oral region. Recently, the Bio-3D printer "Regenova®" has been introduced as an innovative three-dimensional culture system, equipped scaffold-free bio-assembling techniques without any biomaterials. Therefore, we expected a mount of bone defect could be repaired by the structure established from this Bio-3D printer using osteogenic potential stem cells. Material & methods The gingival tissue (1x1 mm) was removed from the distal part of the lower wisdom tooth of the patients who agreed our study. Human Gingival Mesenchymal Stem Cells (hGMSCs) were isolated from this tissue and cultured, since we confirmed the characteristics such as facile isolation and accelerated proliferation, further, strong potential of osteogenic-differentiation. Spheroids were formed using hGMSC in 96-well plates designed for low cell adhesion. The size of the spheroids was measured, and fluorescent immunostaining was employed to verify the expression of stem cell and apoptosis marker, and extracellular matrix. Following four weeks of bone differentiation, μCT imaging was performed. Calcification was confirmed by alizarin red and von Kossa staining. Fluorescent immunostaining was utilized to assess the expression of markers indicative of advanced bone differentiation. Results We have established and confirmed the spheroids (∼600 μm in diameter) constructed from human GMSCs (hGMSCs) still maintain stem cell potentials and osteogenic differentiation abilities from the results that CD73 and not CD34 were expressed as stem cell positive and negative marker, respectively. These spheroids were pilled up like cylindal shape to the "Kenzan" platform of Bio-3D printer and cultured for 7days. The cylindal structure originated from compound spheroids were tried to differentiate into bone four weeks with osteogenic induction medium. The calcification of bio-3D printed bone-like structures was confirmed by alizarin red and Von Kossa staining. In addition, μCT analysis revealed that the HU (Hounsfield Unit) of the calcified structures was almost identical to that of trabecular bone. Immunofluorescent staining detected osteocalcin expression, a late-stage bone differentiation marker. Conclusion For the first time, we have achieved the construction of a scaffold-free, bone-like luminal structure through the assembly of spheroids comprised of this hGMSCs. This success is sure to be close to the induction of clinical application against regenerative medicine especially for bone defect disease.
Collapse
Affiliation(s)
- Masaaki Toyoda
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Takao Fukuda
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Ryota Fujimoto
- Center for Regenerative Medicine Research, Faculty of Medicine, Saga University, Saga, Japan
| | - Kentaro Kawakami
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Chikako Hayashi
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yuki Nakao
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yukari Watanabe
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Tsukasa Aoki
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Miyu Shida
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Terukazu Sanui
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Masahide Taguchi
- Section of Pediatric Dentistry, Department of Oral Growth and Development, Fukuoka Dental College, Fukuoka, Japan
| | - Kensuke Yamamichi
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Ayami Okabe
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tatsunori Okada
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kyoko Oka
- Section of Pediatric Dentistry, Department of Oral Growth and Development, Fukuoka Dental College, Fukuoka, Japan
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
| | - Koichi Nakayama
- Center for Regenerative Medicine Research, Faculty of Medicine, Saga University, Saga, Japan
| | - Fusanori Nishimura
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Shunichi Kajioka
- Department of Pharmaceutical Sciences, International University of Health and Welfare, Fukuoka, Japan
| |
Collapse
|
8
|
Lau CS, Park SY, Ethiraj LP, Singh P, Raj G, Quek J, Prasadh S, Choo Y, Goh BT. Role of Adipose-Derived Mesenchymal Stem Cells in Bone Regeneration. Int J Mol Sci 2024; 25:6805. [PMID: 38928517 PMCID: PMC11204188 DOI: 10.3390/ijms25126805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Bone regeneration involves multiple factors such as tissue interactions, an inflammatory response, and vessel formation. In the event of diseases, old age, lifestyle, or trauma, bone regeneration can be impaired which could result in a prolonged healing duration or requiring an external intervention for repair. Currently, bone grafts hold the golden standard for bone regeneration. However, several limitations hinder its clinical applications, e.g., donor site morbidity, an insufficient tissue volume, and uncertain post-operative outcomes. Bone tissue engineering, involving stem cells seeded onto scaffolds, has thus been a promising treatment alternative for bone regeneration. Adipose-derived mesenchymal stem cells (AD-MSCs) are known to hold therapeutic value for the treatment of various clinical conditions and have displayed feasibility and significant effectiveness due to their ease of isolation, non-invasive, abundance in quantity, and osteogenic capacity. Notably, in vitro studies showed AD-MSCs holding a high proliferation capacity, multi-differentiation potential through the release of a variety of factors, and extracellular vesicles, allowing them to repair damaged tissues. In vivo and clinical studies showed AD-MSCs favoring better vascularization and the integration of the scaffolds, while the presence of scaffolds has enhanced the osteogenesis potential of AD-MSCs, thus yielding optimal bone formation outcomes. Effective bone regeneration requires the interplay of both AD-MSCs and scaffolds (material, pore size) to improve the osteogenic and vasculogenic capacity. This review presents the advances and applications of AD-MSCs for bone regeneration and bone tissue engineering, focusing on the in vitro, in vivo, and clinical studies involving AD-MSCs for bone tissue engineering.
Collapse
Affiliation(s)
- Chau Sang Lau
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - So Yeon Park
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
| | - Lalith Prabha Ethiraj
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Priti Singh
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
| | - Grace Raj
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
| | - Jolene Quek
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (Y.C.)
| | - Somasundaram Prasadh
- Center for Clean Energy Engineering, University of Connecticut, Storrs, CT 06269, USA;
| | - Yen Choo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (Y.C.)
| | - Bee Tin Goh
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| |
Collapse
|
9
|
Ruhl T, Sessler TM, Keimes JM, Beier JP, Villwock S, Rose M, Dahl E. ITIH5 inhibits proliferation, adipogenic differentiation, and secretion of inflammatory cytokines of human adipose stem cells-A new key in treating obesity? FASEB J 2024; 38:e23352. [PMID: 38095340 DOI: 10.1096/fj.202301366r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/26/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023]
Abstract
Inter-α-trypsin inhibitor heavy chain 5 (ITIH5) is widely expressed in the human body, and it is detected to be particularly abundant in adipose tissue. ITIH5 expression is increased in people with obesity compared to lean persons and is decreased by diet-induced weight loss. This suggests that ITIH5 may be involved in the development of adiposity and clinical metabolic variables, although its exact function remains unknown. We measured the protein concentration of ITIH5 in adipose samples from patients undergoing abdominoplasty and tested for correlation with the subjects' BMI as well as inflammatory mediators. We stimulated human adipose stem cells (ASCs) with recombinant (r)ITIH5 protein and tested for an effect on proliferation, differentiation, and immunosuppressive properties when the cells were exposed to an artificial inflammatory environment. We found positive correlations between ITIH5 levels and the BMI (p < .001) as well as concentrations of inflammatory cytokines (TNF-α, IL-6, and MCP-1) in adipose tissue (p < .01). Application of the rITIH5 protein inhibited both proliferation (p < .001) and differentiation of ASCs. Especially, the development of mature adipocytes was reduced by over 50%. Moreover, rITIH5 decreased the release of IL-6 and MCP-1 when the cells were exposed to TNF-α and IL-1β (p < .001). Our data suggest that ITIH5 is an adipokine that is increasingly released during human adipose tissue development, acting as a regulator that inhibits proliferation and adipogenic differentiation of ASCs. ITIH5 thus presents itself as a positive regulator of adipose tissue homeostasis, possibly protecting against both hyperplasia and hypertrophy of adipose tissue and the associated chronic inflammation.
Collapse
Affiliation(s)
- Tim Ruhl
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Thomas M Sessler
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Jana M Keimes
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Justus P Beier
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Sophia Villwock
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Michael Rose
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Edgar Dahl
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
10
|
Assid E, Renshaw A, Samad M, Tupler R, Jones D. Long-Term Outcomes of Osteochondral Allograft with Osteogenic Protein-1 Augmentation: A Twelve-Year Follow-Up. Case Rep Orthop 2023; 2023:3842443. [PMID: 37789911 PMCID: PMC10545460 DOI: 10.1155/2023/3842443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/25/2023] [Accepted: 09/01/2023] [Indexed: 10/05/2023] Open
Abstract
Background Osteochondral lesions (OCLs) can significantly impact functional status and activities of daily living. Weightbearing joints are disproportionately affected due to considerable biomechanical forces in these areas. Various biologic reconstructive procedures such as microfracture, osteochondral autograft transfer (OATS) or allograft transplantation (OCA), and matrix-induced autologous chondrocyte implantation (MACI) are utilized by surgeons to treat OCLs. The integration of osteochondral allografts can restore knee function and maintain the integrity of adjacent joint surfaces. Bone incorporation has been linked to successful outcomes following OCA. Pulse lavage and carbon dioxide have been used to remove marrow elements from the superficial, middle, and deep layers of the allograft; this has been combined with the use of various biologics such as bone marrow aspirate or whole blood to augment donor bone incorporation into the host bone. We present an innovative augmentation approach in OCA transplantation demonstrating excellent incorporation of an osteogenic protein-1 (OP-1) implant (Stryker, Kalamazoo, MI) to treat a large fresh osteoarticular allograft. Case Presentation. We present a 51-year-old male who received OCA augmented with an OP-1 implant (Stryker, Kalamazoo, MI) in 2011. Due to subsequent ACL reconstruction for two years and medial meniscal repair four years following OCA transplantation, we were able to arthroscopically evaluate graft status at short- and intermediate-term follow-ups. Positive findings were further verified with radiographic imaging and patient-reported outcome measures (PROMs). Conclusion OP-1 implants aided in the bone incorporation of a large osteochondral allograft, restoring a high functional level in a demanding sport.
Collapse
Affiliation(s)
- Eric Assid
- Ochsner Health System, New Orleans, LA, USA
| | | | - Mawadah Samad
- The University of Queensland, Brisbane, Queensland, Australia
| | | | | |
Collapse
|
11
|
Thamnium S, Laomeephol C, Pavasant P, Osathanon T, Tabata Y, Wang C, Luckanagul JA. Osteogenic induction of asiatic acid derivatives in human periodontal ligament stem cells. Sci Rep 2023; 13:14102. [PMID: 37644086 PMCID: PMC10465493 DOI: 10.1038/s41598-023-41388-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023] Open
Abstract
Asiatic acid (AA) and asiaticoside, pentacyclic triterpenoid compounds derived from Centella asiatica, are known for their biological effects in promoting type I collagen synthesis and inducing osteogenesis of stem cells. However, their applications in regenerative medicine are limited due to their low potency and poor aqueous solubility. This work aimed to evaluate the osteogenic induction activity of AA derivatives in human periodontal ligament stem cells (hPDLSCs) in vitro. Four compounds were synthesised, namely 501, 502, 503, and 506. AA was used as the control. The 502 exhibited low water solubility, while the 506 compound showed the highest. The cytotoxicity analysis demonstrated that 503 caused significant deterioration in cell viability, while other derivatives showed no harmful effect on hPDLSCs. The dimethyl aminopropyl amine derivative of AA, compound 506, demonstrated a relatively high potency in inducing osteogenic differentiation. An elevated mRNA expression of osteogenic-related genes, BMP2, WNT3A, ALP, OSX and IBSP was observed with 506. Additionally, the expression of BMP-2 protein was enhanced with increasing dose of 506, and the effect was pronounced when the Erk signalling molecule was inhibited. The 506 derivative was proposed for the promotion of osteogenic differentiation in hPDLSCs by upregulating BMP2 via the Erk signalling pathway. The 506 molecule showed promise in bone tissue regeneration.
Collapse
Affiliation(s)
- Sirikool Thamnium
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Chavee Laomeephol
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Biomaterial Engineering in Medical and Health, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Prasit Pavasant
- Center of Excellence in Regenerative Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Thanaphum Osathanon
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Institute for Life and Medical Sciences, Kyoto University, 53 Kawara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Chao Wang
- Chengdu Institute of Biology, Chinese Academy of Sciences, P.O. Box 416, Chengdu, 6100641, Sichuan, People's Republic of China
- School of Food and Bioengineering, Xihua University, Chengdu, 610039, People's Republic of China
| | - Jittima A Luckanagul
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
- Center of Excellence in Biomaterial Engineering in Medical and Health, Chulalongkorn University, Bangkok, 10330, Thailand.
- Center of Excellence in Plant-Produced Pharmaceuticals, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
12
|
Ohnishi T, Homan K, Fukushima A, Ukeba D, Iwasaki N, Sudo H. A Review: Methodologies to Promote the Differentiation of Mesenchymal Stem Cells for the Regeneration of Intervertebral Disc Cells Following Intervertebral Disc Degeneration. Cells 2023; 12:2161. [PMID: 37681893 PMCID: PMC10486900 DOI: 10.3390/cells12172161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/24/2023] [Accepted: 08/26/2023] [Indexed: 09/09/2023] Open
Abstract
Intervertebral disc (IVD) degeneration (IDD), a highly prevalent pathological condition worldwide, is widely associated with back pain. Treatments available compensate for the impaired function of the degenerated IVD but typically have incomplete resolutions because of their adverse complications. Therefore, fundamental regenerative treatments need exploration. Mesenchymal stem cell (MSC) therapy has been recognized as a mainstream research objective by the World Health Organization and was consequently studied by various research groups. Implanted MSCs exert anti-inflammatory, anti-apoptotic, and anti-pyroptotic effects and promote extracellular component production, as well as differentiation into IVD cells themselves. Hence, the ultimate goal of MSC therapy is to recover IVD cells and consequently regenerate the extracellular matrix of degenerated IVDs. Notably, in addition to MSC implantation, healthy nucleus pulposus (NP) cells (NPCs) have been implanted to regenerate NP, which is currently undergoing clinical trials. NPC-derived exosomes have been investigated for their ability to differentiate MSCs from NPC-like phenotypes. A stable and economical source of IVD cells may include allogeneic MSCs from the cell bank for differentiation into IVD cells. Therefore, multiple alternative therapeutic options should be considered if a refined protocol for the differentiation of MSCs into IVD cells is established. In this study, we comprehensively reviewed the molecules, scaffolds, and environmental factors that facilitate the differentiation of MSCs into IVD cells for regenerative therapies for IDD.
Collapse
Affiliation(s)
- Takashi Ohnishi
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan; (T.O.); (K.H.); (A.F.); (N.I.)
| | - Kentaro Homan
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan; (T.O.); (K.H.); (A.F.); (N.I.)
| | - Akira Fukushima
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan; (T.O.); (K.H.); (A.F.); (N.I.)
| | - Daisuke Ukeba
- Department of Orthopedic Surgery, Hokkaido University Hospital, Sapporo 060-8648, Japan;
| | - Norimasa Iwasaki
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan; (T.O.); (K.H.); (A.F.); (N.I.)
| | - Hideki Sudo
- Department of Advanced Medicine for Spine and Spinal Cord Disorders, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| |
Collapse
|
13
|
Vukovic M, Lazarevic M, Mitic D, Jaksic Karisik M, Ilic B, Andric M, Jevtic B, Roganovic J, Milasin J. Acetylsalicylic-acid (ASA) regulation of osteo/odontogenic differentiation and proliferation of human dental pulp stem cells (DPSCs) in vitro. Arch Oral Biol 2022; 144:105564. [PMID: 36215814 DOI: 10.1016/j.archoralbio.2022.105564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/28/2022] [Accepted: 09/29/2022] [Indexed: 11/02/2022]
Abstract
OBJECTIVE The study aimed to investigate acetylsalicylic acid (ASA) effects on osteo/odontogenic differentiation and proliferation of dental pulp stem cells (DPSCs) in vitro and the potential involvement of adenosine monophosphate-activated protein kinase (AMPK) pathway in these processes. DESIGN DPSCs were isolated from third molars pulp tissues of five patients and grown in osteogenic medium alone or supplemented with ASA. Expression of DPSCs markers was tested by flow-cytometry. Cytotoxicity of ASA at concentrations of 10, 50 and 100 µg/ml was tested by MTT and NR assays. Osteo/odontogenic differentiation was analyzed via alizarin red staining and ALP activity. Quantitative PCR (qPCR) was used for osteo/odontogenic markers' (DSPP, BMP2, BMP4, BSP, OCN and RUNX2) and c-Myc expression analysis. AMPK inhibition of ASA-induced osteo/odontogenesis was tested by qPCR of selected markers (DSPP, OCN and RUNX2). RESULTS Cytotoxicity assays showed that only the highest ASA dose decreased cell viability (89.1 %). The smallest concentration of ASA applied on DPSCs resulted in a remarkable enhancement of osteo/odontogenic differentiation, as judged by increased mineralized nodules' formation, ALP activity and gene expression of analyzed markers (increase between 2 and 30 folds), compared to untreated cells. ASA also increased DPSCs proliferation. Interestingly, AMPK inhibition per se upregulated DSPP, OCN and RUNX2; the gene upregulation was higher when ASA treatment was also included. c-Myc expression level decreased in cultures treated with ASA, indicating undergoing differentiation processes. CONCLUSIONS Low concentrations of ASA (corresponding to the standard use in cardiovascular patients), were shown to stimulate osteo/odontogenic differentiation of dental pulp stem cells.
Collapse
Affiliation(s)
- Mladen Vukovic
- Department of Human Genetics, School of Dental Medicine, University of Belgrade, Serbia
| | - Milos Lazarevic
- Department of Human Genetics, School of Dental Medicine, University of Belgrade, Serbia
| | - Dijana Mitic
- Department of Human Genetics, School of Dental Medicine, University of Belgrade, Serbia
| | - Milica Jaksic Karisik
- Department of Human Genetics, School of Dental Medicine, University of Belgrade, Serbia
| | - Branislav Ilic
- Clinic for Oral Surgery, School of Dental Medicine, University of Belgrade, Serbia
| | - Miroslav Andric
- Clinic for Oral Surgery, School of Dental Medicine, University of Belgrade, Serbia
| | - Bojan Jevtic
- Department of Immunology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Serbia
| | - Jelena Roganovic
- Department of Pharmacology in Dentistry, School of Dental Medicine, University of Belgrade, Serbia
| | - Jelena Milasin
- Department of Human Genetics, School of Dental Medicine, University of Belgrade, Serbia.
| |
Collapse
|
14
|
Lu M, Li M, Luo T, Li Y, Wang M, Xue H, Zhang M, Chen Q. Beta-naphthoflavone increases the differentiation of osteoblasts and suppresses adipogenesis in human adipose derived stem cells involving STAT3 pathway. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00283-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
15
|
Hinkelmann S, Springwald AH, Starke A, Kalwa H, Wölk C, Hacker MC, Schulz-Siegmund M. Microtissues from mesenchymal stem cells and siRNA-loaded cross-linked gelatin microparticles for bone regeneration. Mater Today Bio 2022; 13:100190. [PMID: 34988418 PMCID: PMC8693629 DOI: 10.1016/j.mtbio.2021.100190] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/20/2021] [Accepted: 12/11/2021] [Indexed: 12/13/2022] Open
Abstract
The aim of this study was the evaluation of cross-linked gelatin microparticles (cGM) as substrates for osteogenic cell culture to assemble 3D microtissues and their use as delivery system for siRNA to cells in these assemblies. In a 2D transwell cultivation system, we found that cGM are capable to accumulate calcium ions from the surrounding medium. Such a separation of cGM and SaOS-2 cells consequently led to a suppressed matrix mineral formation in the SaOS-2 culture on the well bottom of the transwell system. Thus, we decided to use cGM as component in 3D microtissues and get a close contact between calcium ion accumulating microparticles and cells to improve matrix mineralization. Gelatin microparticles were cross-linked with a N,N-diethylethylenediamine-derivatized (DEED) maleic anhydride (MA) containing oligo (pentaerythritol diacrylate monostearate-co-N-isopropylacrylamide-co-MA) (oPNMA) and aggregated with SaOS-2 or human mesenchymal stem cells (hMSC) to microtissue spheroids. We systematically varied the content of cGM in microtissues and observed cell differentiation and tissue formation. Microtissues were characterized by gene expression, ALP activity and matrix mineralization. Mineralization was detectable in microtissues with SaOS-2 cells after 7 days and with hMSC after 24–28 days in osteogenic culture. When we transfected hMSC via cGM loaded with Lipofectamine complexed chordin siRNA, we found increased ALP activity and accelerated mineral formation in microtissues in presence of BMP-2. As a model for positive paracrine effects that indicate promising in vivo effects of these microtissues, we incubated pre-differentiated microtissues with freshly seeded hMSC monolayers and found improved mineral formation all over the well in the co-culture model. These findings may support the concept of microtissues from hMSC and siRNA-loaded cGM for bone regeneration.
Collapse
Affiliation(s)
- Sandra Hinkelmann
- Institute of Pharmacy, Pharmaceutical Technology, Faculty of Medicine, University of Leipzig, Germany
| | - Alexandra H Springwald
- Institute of Pharmacy, Pharmaceutical Technology, Faculty of Medicine, University of Leipzig, Germany
| | - Annett Starke
- Institute of Pharmacy, Pharmaceutical Technology, Faculty of Medicine, University of Leipzig, Germany
| | - Hermann Kalwa
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Christian Wölk
- Institute of Pharmacy, Pharmaceutical Technology, Faculty of Medicine, University of Leipzig, Germany
| | - Michael C Hacker
- Institute of Pharmacy, Pharmaceutical Technology, Faculty of Medicine, University of Leipzig, Germany.,Institute of Pharmaceutics and Biopharmaceutics, Heinrich Heine University, Düsseldorf, Germany
| | - Michaela Schulz-Siegmund
- Institute of Pharmacy, Pharmaceutical Technology, Faculty of Medicine, University of Leipzig, Germany
| |
Collapse
|
16
|
Xu W, Li Y, Feng R, He P, Zhang Y. γ-Tocotrienol induced the proliferation and differentiation of MC3T3-E1 cells through the stimulation of the Wnt/β-catenin signaling pathway. Food Funct 2022; 13:398-410. [PMID: 34908071 DOI: 10.1039/d1fo02583j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
γ-Tocotrienol (γ-T3), an isoprenoid phytochemical, has shown the promotion of osteoblast proliferation and differentiation in our previous study. In this study, its underlying mechanism was investigated through regulating the Wnt/β-catenin signaling pathway in MC3T3-E1 cells. Comparative experiment results showed that γ-T3, not α-tocopherol (α-TOC) increased more significantly the viability and differentiation in MC3T3-E1 cells. After that, the cells were incubated with 10 mM LiCl, or 4 μM γ-T3 with or without 1 μM XAV-939. γ-T3 at 4 μM stimulated the Wnt/β-catenin signaling pathway by increasing the expression and nuclear accumulation of β-catenin, and the expressions of their downstream factors, such as cyclin-D1, c-Myc, BMP2 and BMP-4 in MC3T3-E1 cells. γ-T3 not only upregulated the viability, induced G0/G1 to the S phase, and promoted the expressions of PCNA (Proliferating Cell Nuclear Antigen) and Ki-67, but also increased ALP activity and the expressions of ON, OPN and OCN. Moreover, the effects of γ-T3 on the MC3T3-E1 cells resembled the actions of LiCl, an activator of the Wnt/β-catenin signaling pathway. Notably, all these effects of γ-T3 on the MC3T3-E1 cells were completely blocked by the Wnt/β-catenin signaling pathway inhibitor XAV-939. Our data demonstrated that γ-T3 can target β-catenin to enhance the Wnt/β-catenin signaling pathway, which led to increased expressions of the downstream cell proliferation and cell cycle-associated (cyclin D1 and c-myc), and cell differentiation-associated (BMP-2 and BMP-4) target genes, and ultimately promoted MC3T3-E1 cell proliferation and differentiation. Therefore, γ-T3 may be a potential agent to prevent and reverse osteoporosis due to its safety and powerful abilities of osteogenesis.
Collapse
Affiliation(s)
- Weili Xu
- Innovation Research Center for Special Food-Medicine and Biochemical Engineering, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, 92 Xidazhi Street, Nangang District, Harbin, China.
| | - Yutong Li
- Innovation Research Center for Special Food-Medicine and Biochemical Engineering, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, 92 Xidazhi Street, Nangang District, Harbin, China.
| | - Rennan Feng
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, China
| | - Pan He
- Innovation Research Center for Special Food-Medicine and Biochemical Engineering, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, 92 Xidazhi Street, Nangang District, Harbin, China.
| | - Yuqi Zhang
- Innovation Research Center for Special Food-Medicine and Biochemical Engineering, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, 92 Xidazhi Street, Nangang District, Harbin, China.
| |
Collapse
|
17
|
Doyle SE, Snow F, Duchi S, O’Connell CD, Onofrillo C, Di Bella C, Pirogova E. 3D Printed Multiphasic Scaffolds for Osteochondral Repair: Challenges and Opportunities. Int J Mol Sci 2021; 22:12420. [PMID: 34830302 PMCID: PMC8622524 DOI: 10.3390/ijms222212420] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 12/19/2022] Open
Abstract
Osteochondral (OC) defects are debilitating joint injuries characterized by the loss of full thickness articular cartilage along with the underlying calcified cartilage through to the subchondral bone. While current surgical treatments can provide some relief from pain, none can fully repair all the components of the OC unit and restore its native function. Engineering OC tissue is challenging due to the presence of the three distinct tissue regions. Recent advances in additive manufacturing provide unprecedented control over the internal microstructure of bioscaffolds, the patterning of growth factors and the encapsulation of potentially regenerative cells. These developments are ushering in a new paradigm of 'multiphasic' scaffold designs in which the optimal micro-environment for each tissue region is individually crafted. Although the adoption of these techniques provides new opportunities in OC research, it also introduces challenges, such as creating tissue interfaces, integrating multiple fabrication techniques and co-culturing different cells within the same construct. This review captures the considerations and capabilities in developing 3D printed OC scaffolds, including materials, fabrication techniques, mechanical function, biological components and design.
Collapse
Affiliation(s)
- Stephanie E. Doyle
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia; (F.S.)
- ACMD, St Vincent’s Hospital Melbourne, Fitzroy, VIC 3065, Australia; (S.D.); (C.O.); (C.D.B.)
| | - Finn Snow
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia; (F.S.)
| | - Serena Duchi
- ACMD, St Vincent’s Hospital Melbourne, Fitzroy, VIC 3065, Australia; (S.D.); (C.O.); (C.D.B.)
- Department of Surgery, The University of Melbourne, St Vincent’s Hospital Melbourne, Fitzroy, VIC 3065, Australia
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Cathal D. O’Connell
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia; (F.S.)
- ACMD, St Vincent’s Hospital Melbourne, Fitzroy, VIC 3065, Australia; (S.D.); (C.O.); (C.D.B.)
| | - Carmine Onofrillo
- ACMD, St Vincent’s Hospital Melbourne, Fitzroy, VIC 3065, Australia; (S.D.); (C.O.); (C.D.B.)
- Department of Surgery, The University of Melbourne, St Vincent’s Hospital Melbourne, Fitzroy, VIC 3065, Australia
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Claudia Di Bella
- ACMD, St Vincent’s Hospital Melbourne, Fitzroy, VIC 3065, Australia; (S.D.); (C.O.); (C.D.B.)
- Department of Surgery, The University of Melbourne, St Vincent’s Hospital Melbourne, Fitzroy, VIC 3065, Australia
- Department of Orthopaedics, St Vincent’s Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Elena Pirogova
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia; (F.S.)
| |
Collapse
|
18
|
Hu T, Liu L, Lam RWM, Toh SY, Abbah SA, Wang M, Ramruttun AK, Bhakoo K, Cool S, Li J, Cho-Hong Goh J, Wong HK. Bone marrow mesenchymal stem cells with low dose bone morphogenetic protein 2 enhances scaffold-based spinal fusion in a porcine model. J Tissue Eng Regen Med 2021; 16:63-75. [PMID: 34687157 DOI: 10.1002/term.3260] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/20/2021] [Accepted: 10/15/2021] [Indexed: 11/07/2022]
Abstract
High doses bone morphogenetic protein 2 (BMP-2) have resulted in a series of complications in spinal fusion. We previously established a polyelectrolyte complex (PEC) carrier system that reduces the therapeutic dose of BMP-2 in both rodent and porcine spinal fusion models. This study aimed to evaluate the safety and efficacy of the combination of bone marrow mesenchymal stem cells (BMSCs) and low dose BMP-2 delivered by PEC for bone regeneration in a porcine model of anterior lumbar interbody spinal fusion (ALIF) application. Six Yorkshire pigs underwent a tri-segmental (L2/L3; L3/L4; L4/L5) ALIF in four groups, namely: (a) BMSCs + 25 μg BMP-2/PEC (n = 9), (b) 25 μg BMP-2/PEC (n = 3), (c) BMSCs (n = 3), and (d) 50 μg BMP-2/absorbable collagen sponge (n = 3). Fusion outcomes were evaluated by radiography, biomechanical testing, and histological analysis after 12 weeks. Mean radiographic scores at 12 weeks were 2.7, 2.0, 1.0, and 1.0 for Groups 1 to 4, respectively. μ-CT scanning, biomechanical evaluation, and histological analysis demonstrated solid fusion and successful bone regeneration in Group 1. In contrast, Group 2 showed inferior quality and slow rate of fusion, and Groups 3 and 4 failed to fuse any of the interbody spaces. There was no obvious evidence of seroma formation, implant rejection, or any other complications in all groups. The results suggest that the combination of BMSCs and low dose BMP-2/PEC could further lower down the effective dose of the BMP-2 and be used as a bone graft substitute in the large animal ALIF model.
Collapse
Affiliation(s)
- Tao Hu
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Spine Surgery, Tongji University School of Medicine, Shanghai East Hospital, Shanghai, China
| | - Ling Liu
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore
| | - Raymond Wing Moon Lam
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Soo Yein Toh
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sunny Akogwu Abbah
- Department of Obstetrics and Gynaecology, Portiuncula University Hospital Ballinasloe, Galway, Ireland.,CÚRAM, Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
| | - Ming Wang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Amit Kumarsing Ramruttun
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kishore Bhakoo
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Simon Cool
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jun Li
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore
| | - James Cho-Hong Goh
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore
| | - Hee-Kit Wong
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Tissue Engineering Programme (NUSTEP), Life Sciences Institute, Singapore, Singapore
| |
Collapse
|
19
|
Microenvironment Influences on Human Umbilical Cord Mesenchymal Stem Cell-Based Bone Regeneration. Stem Cells Int 2021; 2021:4465022. [PMID: 34447439 PMCID: PMC8384552 DOI: 10.1155/2021/4465022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/26/2021] [Indexed: 01/08/2023] Open
Abstract
The microenvironment, or niche, regulates stem cell fate and improves differentiation efficiency. Human umbilical cord mesenchymal stem cells (hUC-MSCs) are ideal cell source for bone tissue engineering. However, the role of the microenvironments in hUC-MSC-based bone regeneration is not yet fully understood. This study is aimed at investigating the effects of the in vitro culture microenvironment (hUC-MSCs, nano-hydroxyapatite/collagen/poly (L-lactide) (nHAC/PLA), osteogenic media (OMD), and recombinant human bone morphogenetic protein-7 (rhBMP-7)) and the in vivo transplanted microenvironment (ectopic and orthotopic) on bone regeneration ability of hUC-MSCs. The isolated hUC-MSCs showed self-renewal potential and MSCs' characteristics. In the in vitro two-dimensional culture microenvironment, OMD or OMD with rhBMP-7 significantly enhanced hUC-MSCs' osteocalcin immunofluorescence staining, alkaline phosphatase, and Alizarin red staining; OMD with rhBMP-7 exhibited the highest ALP secretion and mineralized matrix formation. In the in vitro three-dimensional culture microenvironment, nHAC/PLA supported hUC-MSCs' adhesion, proliferation, and differentiation; the microenvironment containing OMD or OMD and rhBMP-7 shortened cell proliferation progression and made osteogenic differentiation progression advance; rhBMP-7 significantly attenuated the inhibiting effect of OMD on hUC-MSCs' proliferation and significantly enhanced the promoting effect of OMD on gene expression and protein secretion of osteogenic differentiation markers, calcium and phosphorous concentration, and mineralized matrix formation. The in vitro three-dimensional culture microenvironment containing OMD and rhBMP-7 induced hUC-MSCs to form the most new bones in ectopic or orthotopic microenvironment as proved by microcomputed tomography and hematoxylin and eosin staining, but bone formation in orthotopic microenvironment was significantly higher than that in ectopic microenvironment. The results indicated that the combination of in vitro hUC-MSCs+nHAC/PLA+OMD+rhBMP-7 microenvironment and in vivo orthotopic microenvironment provided a more optimized niche for bone regeneration of hUC-MSCs. This study elucidates that hUC-MSCs and their local microenvironment, or niche, play an important role in hUC-MSC-based bone regeneration. The endogenously produced BMP may serve an important regulatory role in the process.
Collapse
|
20
|
Bjelić D, Finšgar M. The Role of Growth Factors in Bioactive Coatings. Pharmaceutics 2021; 13:1083. [PMID: 34371775 PMCID: PMC8309025 DOI: 10.3390/pharmaceutics13071083] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 12/26/2022] Open
Abstract
With increasing obesity and an ageing population, health complications are also on the rise, such as the need to replace a joint with an artificial one. In both humans and animals, the integration of the implant is crucial, and bioactive coatings play an important role in bone tissue engineering. Since bone tissue engineering is about designing an implant that maximally mimics natural bone and is accepted by the tissue, the search for optimal materials and therapeutic agents and their concentrations is increasing. The incorporation of growth factors (GFs) in a bioactive coating represents a novel approach in bone tissue engineering, in which osteoinduction is enhanced in order to create the optimal conditions for the bone healing process, which crucially affects implant fixation. For the application of GFs in coatings and their implementation in clinical practice, factors such as the choice of one or more GFs, their concentration, the coating material, the method of incorporation, and the implant material must be considered to achieve the desired controlled release. Therefore, the avoidance of revision surgery also depends on the success of the design of the most appropriate bioactive coating. This overview considers the integration of the most common GFs that have been investigated in in vitro and in vivo studies, as well as in human clinical trials, with the aim of applying them in bioactive coatings. An overview of the main therapeutic agents that can stimulate cells to express the GFs necessary for bone tissue development is also provided. The main objective is to present the advantages and disadvantages of the GFs that have shown promise for inclusion in bioactive coatings according to the results of numerous studies.
Collapse
Affiliation(s)
| | - Matjaž Finšgar
- Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia;
| |
Collapse
|
21
|
Nourisa J, Zeller-Plumhoff B, Helmholz H, Luthringer-Feyerabend B, Ivannikov V, Willumeit-Römer R. Magnesium ions regulate mesenchymal stem cells population and osteogenic differentiation: A fuzzy agent-based modeling approach. Comput Struct Biotechnol J 2021; 19:4110-4122. [PMID: 34527185 PMCID: PMC8346546 DOI: 10.1016/j.csbj.2021.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are proliferative and multipotent cells that play a key role in the bone regeneration process. Empirical data have repeatedly shown the bioregulatory importance of magnesium (Mg) ions in MSC growth and osteogenesis. In this study, we propose an agent-based model to predict the spatiotemporal dynamics of the MSC population and osteogenic differentiation in response to Mg2+ ions. A fuzzy-logic controller was designed to govern the decision-making process of cells by predicting four cellular processes of proliferation, differentiation, migration, and mortality in response to several important bioregulatory factors such as Mg2+ ions, pH, BMP2, and TGF-β1. The model was calibrated using the empirical data obtained from three sets of cell culture experiments. The model successfully reproduced the empirical observations regarding live cell count, viability, DNA content, and the differentiation-related markers of alkaline phosphate (ALP) and osteocalcin (OC). The simulation results, in agreement with the empirical data, showed that Mg2+ ions within 3-6 mM concentration have the highest stimulation effect on cell population growth. The model also correctly reproduced the stimulatory effect of Mg2+ ions on ALP and its inhibitory effect on OC as the early and late differentiation markers, respectively. Besides, the numerical simulation shed light on the innate cellular differences of the cells cultured in different experiments in terms of the proliferative capacity as well as sensitivity to Mg2+ ions. The proposed model can be adopted in the study of the osteogenesis around Mg-based implants where ions released due to degradation interact with local cells and regulate bone regeneration.
Collapse
Affiliation(s)
- Jalil Nourisa
- Helmholtz Zentrum Hereon, Institute of Metallic Biomaterials, Max-Planck-Straße 1, 21502 Geesthacht, Germany
| | - Berit Zeller-Plumhoff
- Helmholtz Zentrum Hereon, Institute of Metallic Biomaterials, Max-Planck-Straße 1, 21502 Geesthacht, Germany
| | - Heike Helmholz
- Helmholtz Zentrum Hereon, Institute of Metallic Biomaterials, Max-Planck-Straße 1, 21502 Geesthacht, Germany
| | | | - Vladimir Ivannikov
- Helmholtz Zentrum Hereon, Institute of Metallic Biomaterials, Max-Planck-Straße 1, 21502 Geesthacht, Germany
| | - Regine Willumeit-Römer
- Helmholtz Zentrum Hereon, Institute of Metallic Biomaterials, Max-Planck-Straße 1, 21502 Geesthacht, Germany
| |
Collapse
|
22
|
Borgiani E, Duda GN, Willie BM, Checa S. Bone morphogenetic protein 2-induced cellular chemotaxis drives tissue patterning during critical-sized bone defect healing: an in silico study. Biomech Model Mechanobiol 2021; 20:1627-1644. [PMID: 34047890 PMCID: PMC8298257 DOI: 10.1007/s10237-021-01466-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/11/2021] [Indexed: 12/26/2022]
Abstract
Critical-sized bone defects are critical healing conditions that, if left untreated, often lead to non-unions. To reduce the risk, critical-sized bone defects are often treated with recombinant human BMP-2. Although enhanced bone tissue formation is observed when BMP-2 is administered locally to the defect, spatial and temporal distribution of callus tissue often differs from that found during regular bone healing or in defects treated differently. How this altered tissue patterning due to BMP-2 treatment is linked to mechano-biological principles at the cellular scale remains largely unknown. In this study, the mechano-biological regulation of BMP-2-treated critical-sized bone defect healing was investigated using a multiphysics multiscale in silico approach. Finite element and agent-based modeling techniques were combined to simulate healing within a critical-sized bone defect (5 mm) in a rat femur. Computer model predictions were compared to in vivo microCT data outcome of bone tissue patterning at 2, 4, and 6 weeks postoperation. In vivo, BMP-2 treatment led to complete healing through periosteal bone bridging already after 2 weeks postoperation. Computer model simulations showed that the BMP-2 specific tissue patterning can be explained by the migration of mesenchymal stromal cells to regions with a specific concentration of BMP-2 (chemotaxis). This study shows how computational modeling can help us to further understand the mechanisms behind treatment effects on compromised healing conditions as well as to optimize future treatment strategies.
Collapse
Affiliation(s)
- Edoardo Borgiani
- Julius Wolff Institute, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Institutsgebäude Süd/ Südstraße 2, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Georg N Duda
- Julius Wolff Institute, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Institutsgebäude Süd/ Südstraße 2, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Bettina M Willie
- Research Centre, Department of Pediatric Surgery, Shriners Hospital for Children-Canada, McGill University, 1003 Decarie Blvd, Montreal, QC, H4A 0A9, Canada
| | - Sara Checa
- Julius Wolff Institute, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Institutsgebäude Süd/ Südstraße 2, Augustenburger Platz 1, 13353, Berlin, Germany.
| |
Collapse
|
23
|
Zhang X, He J, Wang W. Progress in the use of mesenchymal stromal cells for osteoarthritis treatment. Cytotherapy 2021; 23:459-470. [PMID: 33736933 DOI: 10.1016/j.jcyt.2021.01.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 12/20/2020] [Accepted: 01/29/2021] [Indexed: 12/26/2022]
Abstract
LITERATURE REVIEW OF MSCS IN THE TREATMENT OF OSTEOARTHRITIS IN THE PAST FIVE YEARS: Osteoarthritis (OA) is one of the most common chronic joint diseases, with prominent symptoms caused by many factors. However, current medical interventions for OA have resulted in poor clinical outcomes, demonstrating that there are huge unmet medical needs in this area. Cell therapy has opened new avenues of OA treatment. Different sources of mesenchymal stromal cells (MSCs) may have different phenotypes and cellular functions. Pre-clinical and clinical studies have demonstrated the feasibility, safety and efficacy of MSC therapy. Mitogen-activated protein kinase, Wnt and Notch signaling pathways are involved in the chondrogenesis of MSC-mediated treatments. MSCs may also exert effective immunoregulatory and paracrine effects to stimulate tissue repair. Therapy with extracellular vesicles containing cytokines, which are secreted by MSCs, might be a potential treatment for OA.
Collapse
Affiliation(s)
- Xiaotian Zhang
- Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jiyin He
- Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Wen Wang
- Clinical Development, IASO Biotherapeutics Co., Ltd., Shanghai, China.
| |
Collapse
|
24
|
Khazaei S, Keshavarz G, Bozorgi A, Nazari H, Khazaei M. Adipose tissue-derived stem cells: a comparative review on isolation, culture, and differentiation methods. Cell Tissue Bank 2021; 23:1-16. [PMID: 33616792 DOI: 10.1007/s10561-021-09905-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/01/2021] [Indexed: 01/14/2023]
Abstract
Adipose tissue-derived stem cells (ADSCs) are an available source of mesenchymal stem cells with the appropriate capacity to in vitro survive, propagate, and differentiate into cells from three lineages of ectoderm, mesoderm, and endoderm. The biological features of ADSCs depend on the donor physiology and health status, isolation procedure, culture conditions, and differentiation protocols used. Adipose tissue samples are provided by surgery and lipoaspiration-based methods and subjected to various mechanical and chemical digestion techniques to finally generate a heterogeneous mixture named stromal vascular fraction (SVF). ADSCs are purified through varied cell populations that exist within SVF and cultured under standard conditions to give rise to a highly rich resource of stem cells directly applied in the clinic or differentiated into a wide range of cells. The development and optimization of conventional isolation, expansion, and differentiation methods seem noteworthy to preserve the desirable biological functions of ADSCs in pre-clinical and clinical investigations.
Collapse
Affiliation(s)
- Saber Khazaei
- Department of Endodontics, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ghazal Keshavarz
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Azam Bozorgi
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Nazari
- Department of Orofacial Surgery, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mozafar Khazaei
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
25
|
Dubey S, Mishra R, Roy P, Singh RP. 3-D macro/microporous-nanofibrous bacterial cellulose scaffolds seeded with BMP-2 preconditioned mesenchymal stem cells exhibit remarkable potential for bone tissue engineering. Int J Biol Macromol 2020; 167:934-946. [PMID: 33189758 DOI: 10.1016/j.ijbiomac.2020.11.049] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/30/2020] [Accepted: 11/07/2020] [Indexed: 12/13/2022]
Abstract
Bone repair using BMP-2 is a promising therapeutic approach in clinical practices, however, high dosages required to be effective pose issues of cost and safety. The present study explores the potential of low dose BMP-2 treatment via tissue engineering approach, which amalgamates 3-D macro/microporous-nanofibrous bacterial cellulose (mNBC) scaffolds and low dose BMP-2 primed murine mesenchymal stem cells (C3H10T1/2 cells). Initial studies on cell-scaffold interaction using unprimed C3H10T1/2 cells confirmed that scaffolds provided a propitious environment for cell adhesion, growth, and infiltration, owing to its ECM-mimicking nano-micro-macro architecture. Osteogenic studies were conducted by preconditioning the cells with 50 ng/mL BMP-2 for 15 min, followed by culturing on mNBC scaffolds for up to three weeks. The results showed an early onset and significantly enhanced bone matrix secretion and maturation in the scaffolds seeded with BMP-2 primed cells compared to the unprimed ones. Moreover, mNBC scaffolds alone were able to facilitate the mineralization of cells to some extent. These findings suggest that, with the aid of 'osteoinduction' from low dose BMP-2 priming of stem cells and 'osteoconduction' from nano-macro/micro topography of mNBC scaffolds, a cost-effective bone tissue engineering strategy can be designed for quick and excellent in vivo osseointegration.
Collapse
Affiliation(s)
- Swati Dubey
- Microbial Biotechnology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, India.
| | - Rutusmita Mishra
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Partha Roy
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - R P Singh
- Microbial Biotechnology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, India.
| |
Collapse
|
26
|
Osteoconductive hybrid hyaluronic acid hydrogel patch for effective bone formation. J Control Release 2020; 327:571-583. [DOI: 10.1016/j.jconrel.2020.09.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/01/2020] [Accepted: 09/03/2020] [Indexed: 12/18/2022]
|
27
|
Qin Y, Li G, Wang C, Zhang D, Zhang L, Fang H, Yan S, Zhang K, Yin J. Biomimetic Bilayer Scaffold as an Incubator to Induce Sequential Chondrogenesis and Osteogenesis of Adipose Derived Stem Cells for Construction of Osteochondral Tissue. ACS Biomater Sci Eng 2020; 6:3070-3080. [PMID: 33463252 DOI: 10.1021/acsbiomaterials.0c00200] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Toward osteochondral tissue construction, the present study introduced a bilayer scaffold to induce sequential chondrogenesis and osteogenesis of stem cells in vitro. Two scaffolds that are both based on poly(l-glutamic acid) (PLGA) and chitosan (CS) were combined to form the bilayer scaffold. The cartilage region was the covalently cross-linked PLGA/CS hydrogel with a tubular pore structure, possessing a swollen network to prevent cellular adhesion, while inducing spontaneous cellular aggregate formation. The bone region was the electrostatically cross-linked PLGA-grafted nano hydroxyapatite (nHA-g-PLGA)/CS scaffold, which supported cellular adhesion and spreading. Human adipose derived stem cells (hASCs) were seeded into the cartilage region and observed to aggregate, formimg multicellular spheroids, which subsequently fused to rod-like aggregates with a larger size. At the same time, hASCs in aggregates crossed the interface and entered the bone region, presenting adhesion and spreading. With the induction of bone morphogenetic protein 2 (BMP-2) and insulin-like growth factor 1 (IGF-1) during the first 14 days and BMP-2 alone during the last 14 days, hASCs aggregates in the cartilage region underwent chondrogenesis, expressing an abundant cartilage matrix including glycosaminoglycans (GAGs) and type II collagen (COL II) at 28 days. The chondrogenic induced hASCs migrated in the bone region turned to osteogenesis at 28 days, which was associated with their large spreading area and the switch of the induce factor. Thus, the present bilayer scaffold induced the different distribution of hASCs, resulting in subsequent chondrogenesis and osteogenesis, realizing osteochondral tissue construction in vitro.
Collapse
Affiliation(s)
- Yechi Qin
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, China
| | - Guifei Li
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, China
| | - Chen Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Danqing Zhang
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, China
| | - Lili Zhang
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, China
| | - Haowei Fang
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, China
| | - Shifeng Yan
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, China
| | - Kunxi Zhang
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, China
| | - Jingbo Yin
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, China
| |
Collapse
|
28
|
Tian YI, Zhang X, Torrejon K, Danias J, Gindina S, Nayyar A, Du Y, Xie Y. A bioengineering approach to Schlemm's canal-like stem cell differentiation for in vitro glaucoma drug screening. Acta Biomater 2020; 105:203-213. [PMID: 31982588 DOI: 10.1016/j.actbio.2020.01.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/30/2022]
Abstract
Human Schlemm's canal (HSC) cells are critical for understanding outflow physiology and glaucoma etiology. However, primary donor cells frequently used in research are difficult to isolate. HSC cells exhibit both vascular and lymphatic markers. Human adipose-derived stem cells (ADSCs) represent a potential source of HSC due to their capacity to differentiate into both vascular and lymphatic endothelial cells, via VEGF-A and VEGF-C. Shear stress plays a critical role in maintaining HSC integrity, function, and PROX1 expression. Additionally, the human trabecular meshwork (HTM) microenvironment could provide cues for HSC-like differentiation. We hypothesize that subjecting ADSCs to VEGF-A or VEGF-C, shear stress, and co-culture with HTM cells could provide biological, mechanical, and cellular cues necessary for HSC-like differentiation. To test this hypothesis, effects of VEGF-A, VEGF-C, and shear stress on ADSC differentiation were examined and compared to primary HSC cells in terms of cell morphology, and HSC marker expression using qPCR, immunoblotting, and immunocytochemistry analysis. Furthermore, the effect of co-culture with HTM cells on porous scaffolds on ADSC differentiation was studied. Treatment with VEGF-C under shear stress is effective in differentiating ADSCs into PROX1-expressing HSC-like cells. Co-culture with HTM cells on porous scaffolds leads to HTM/ADSC-derived HSC-like constructs that regulate through-flow and respond as expected to dexamethasone. STATEMENT OF SIGNIFICANCE: We successfully generated human Schlemm's canal (HSC) like cells from adipocyte-derived stem cells induced by biochemical and biomechanical cues as well as bioengineered human trabecular meshwork (HTM) on micropatterned, porous SU8 scaffolds. These stem cell-derived HSC-like cells co-cultured with HTM cells on SU8 scaffolds can regulate through-flow, and in particular, are responsive to steroid treatment as expected. These findings show that ADSC-derived HSC-like cells have the potential to recreate the ocular outflow pathway for in vitro glaucoma drug screening. To the best of our knowledge, it is the very first time to demonstrate derivation of Schlemm's canal-like cells from stem cells. It provides an important alternative source to primary Schlemm's canal cells that are very difficult to be isolated and cultured from human donors.
Collapse
Affiliation(s)
- Yangzi Isabel Tian
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, 257 Fuller Road, Albany, NY 12203, USA
| | - Xulang Zhang
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, 257 Fuller Road, Albany, NY 12203, USA
| | - Karen Torrejon
- Glauconix Biosciences, Inc., 251 Fuller Road, Albany, NY 12203, USA
| | - John Danias
- SUNY Downstate Medical Center, 450 Clarkson Ave, Brooklyn, NY 11203, USA
| | - Sofya Gindina
- SUNY Downstate Medical Center, 450 Clarkson Ave, Brooklyn, NY 11203, USA
| | - Ashima Nayyar
- SUNY Downstate Medical Center, 450 Clarkson Ave, Brooklyn, NY 11203, USA
| | - Yiqin Du
- University of Pittsburg School of Medicine, 203 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Yubing Xie
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, 257 Fuller Road, Albany, NY 12203, USA.
| |
Collapse
|
29
|
Gugjoo MB, Amarpal, Fazili MUR, Shah RA, Saleem Mir M, Sharma GT. Goat mesenchymal stem cell basic research and potential applications. Small Rumin Res 2020. [DOI: 10.1016/j.smallrumres.2019.106045] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
30
|
Marycz K, Smieszek A, Targonska S, Walsh SA, Szustakiewicz K, Wiglusz RJ. Three dimensional (3D) printed polylactic acid with nano-hydroxyapatite doped with europium(III) ions (nHAp/PLLA@Eu 3+) composite for osteochondral defect regeneration and theranostics. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 110:110634. [PMID: 32204070 DOI: 10.1016/j.msec.2020.110634] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 12/31/2019] [Accepted: 01/03/2020] [Indexed: 12/20/2022]
Abstract
In the current research previously developed composites composed from poly (l-lactide) (PLLA) and nano-hydroxyapatite (10 wt% nHAp/PLLA) were functionalized with different concentrations of europium (III) (Eu3+). The aim of this study was to determine whether Eu3+ ions doped within the 10 wt% nHAp/PLLA scaffolds will improve the bioactivity of composites. Therefore, first set of experiments was designed to evaluate the effect of Eu3+ ions on morphology, viability, proliferation and metabolism of progenitor cells isolated from adipose tissue (hASC). Three different concentration were tested i.e. 1 mol%, 3 mol% and 5%mol. We identified the 10 wt% nHAp/PLLA@3 mol% Eu3+ scaffolds as the most cytocompatible. Further, we investigated the influence of the composites doped with 3 mol% Eu3+ ions on differentiation of hASC toward bone and cartilage forming cells. Our results showed that 10 wt% nHAp/PLLA@3 mol% Eu3+ scaffolds promotes osteogenesis and chondrogenesis of hASCs what was associated with improved synthesis and secretion of extracellular matrix proteins specific for bone and articular cartilage tissue. We also proved that obtained biomaterials have bio-imaging function and their integration with bone can be monitored using micro computed tomography (μCT).
Collapse
Affiliation(s)
- Krzysztof Marycz
- University of Environmental and Life Sciences Wroclaw, The Department of Experimental Biology, The Faculty of Biology and Animal Science, 38 C Chelmonskiego St., 50-630 Wroclaw, Poland; Collegium Medicum, Cardinal Stefan Wyszynski University (UKSW), Woycickiego 1/3, 01-938 Warsaw, Poland
| | - Agnieszka Smieszek
- University of Environmental and Life Sciences Wroclaw, The Department of Experimental Biology, The Faculty of Biology and Animal Science, 38 C Chelmonskiego St., 50-630 Wroclaw, Poland
| | - Sara Targonska
- Institute of Low Temperature and Structure Research, Polish Academy of Sciences, Okolna 2, PL-50-422 Wroclaw, Poland
| | - Susan A Walsh
- Small Animal Imaging Core, University of Iowa Carver College of Medicine, Iowa City, IA, United States of America
| | - Konrad Szustakiewicz
- Polymer Engineering and Technology Division, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Rafal J Wiglusz
- Institute of Low Temperature and Structure Research, Polish Academy of Sciences, Okolna 2, PL-50-422 Wroclaw, Poland; Centre for Advanced Materials and Smart Structures, Polish Academy of Sciences, Okolna 2, 50-950 Wroclaw, Poland.
| |
Collapse
|
31
|
Hao M, He J, Wang C, Wang C, Ma B, Zhang S, Duan J, Liu F, Zhang Y, Han L, Liu H, Sang Y. Effect of Hydroxyapatite Nanorods on the Fate of Human Adipose-Derived Stem Cells Assessed In Situ at the Single Cell Level with a High-Throughput, Real-Time Microfluidic Chip. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1905001. [PMID: 31697037 DOI: 10.1002/smll.201905001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/05/2019] [Indexed: 06/10/2023]
Abstract
The fate of stem cells at the single cell level with limited communication with other cells is still unknown due to the lack of an efficient tool for highly accurate molecular detection. Moreover, the conditional sensitivity of biological experiments requires a sufficient number of parallel experiments to support a conclusion. In this work, a microfluidic single cell chip is designed for use with a protein chip to investigate the effect of hydroxyapatite (HAp) on the osteogenic differentiation of human adipose-derived stem cells (hADSCs) in situ at the single cell level. By successfully detecting secretory proteins in situ, it is found that the HAp nanorods enhance osteogenic differentiation at the single cell level. In the chip, the single cell seeding approach confirms the osteogenic differentiation of the hADSCs, which endocytoses HAp, by reducing the influence of the factors secreted by neighboring differentiating cells. Most importantly, more than 7000 microchambers provide a sufficient number of parallel experiments for statistical analysis, which ensure a high level of repeatability of the HAp nanorod-induced osteogenic differentiation. The microfluidic chip comprising single cell culture microchambers with in situ detection capability is a promising tool for research on cell behavior or cell fate at the single cell level.
Collapse
Affiliation(s)
- Min Hao
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, P. R. China
| | - Jianlong He
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, P. R. China
| | - Chunhua Wang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, P. R. China
| | - Chao Wang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, P. R. China
| | - Baojin Ma
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, P. R. China
| | - Shan Zhang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, P. R. China
| | - Jiazhi Duan
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, P. R. China
| | - Feng Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, P. R. China
| | - Yu Zhang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, P. R. China
| | - Lin Han
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, P. R. China
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, P. R. China
- Institute for Advanced Interdisciplinary Research (IAIR), University of Jinan, Jinan, 250022, P. R. China
| | - Yuanhua Sang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, P. R. China
| |
Collapse
|
32
|
Allogeneic Versus Autologous Injectable Mesenchymal Stem Cells for Knee Osteoarthritis: Review and Current Status. Tech Orthop 2019. [DOI: 10.1097/bto.0000000000000357] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
33
|
Padiolleau L, Chanseau C, Durrieu S, Ayela C, Laroche G, Durrieu M. Directing hMSCs fate through geometrical cues and mimetics peptides. J Biomed Mater Res A 2019; 108:201-211. [DOI: 10.1002/jbm.a.36804] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 06/11/2019] [Accepted: 09/16/2019] [Indexed: 12/23/2022]
Affiliation(s)
- Laurence Padiolleau
- Chimie et Biologie des Membranes et Nano‐Objets (UMR5248 CBMN) University Bordeaux Pessac France
- CNRS, CBMN UMR5248 Pessac France
- Bordeaux INP, CBMN UMR5248 Pessac France
- Laboratoire d'Ingénierie de Surface (LIS), Département de Génie des Mines, de la Métallurgie et des Matériaux Centre de Recherche sur les Matériaux Avancés (CERMA), Université Laval Québec Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Québec (CRCHUQ), Hôpital St‐François d'Assise Québec Canada
| | - Christel Chanseau
- Chimie et Biologie des Membranes et Nano‐Objets (UMR5248 CBMN) University Bordeaux Pessac France
- CNRS, CBMN UMR5248 Pessac France
- Bordeaux INP, CBMN UMR5248 Pessac France
| | - Stéphanie Durrieu
- ARNA Laboratory Université de Bordeaux Bordeaux France
- ARNA Laboratory INSERM, U1212 – CNRS UMR 5320 Bordeaux France
| | - Cédric Ayela
- Université de Bordeaux, IMS, UMR CNRS 5218 Talence France
| | - Gaétan Laroche
- Laboratoire d'Ingénierie de Surface (LIS), Département de Génie des Mines, de la Métallurgie et des Matériaux Centre de Recherche sur les Matériaux Avancés (CERMA), Université Laval Québec Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Québec (CRCHUQ), Hôpital St‐François d'Assise Québec Canada
| | - Marie‐Christine Durrieu
- Chimie et Biologie des Membranes et Nano‐Objets (UMR5248 CBMN) University Bordeaux Pessac France
- CNRS, CBMN UMR5248 Pessac France
- Bordeaux INP, CBMN UMR5248 Pessac France
| |
Collapse
|
34
|
Deng B, Zhu W, Duan Y, Hu Y, Chen X, Song S, Yi Z, Song Y. Exendin‑4 promotes osteogenic differentiation of adipose‑derived stem cells and facilitates bone repair. Mol Med Rep 2019; 20:4933-4942. [PMID: 31661134 PMCID: PMC6854547 DOI: 10.3892/mmr.2019.10764] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/10/2019] [Indexed: 02/07/2023] Open
Abstract
Inflammation-related bone defects pose a heavy burden on patients and orthopedic surgeons. Although stem-cell-based bone repair has developed rapidly, it is of great significance to characterize bio-active molecules that facilitate bone regeneration. It is reported that a glucagon-like peptide 1 receptor agonist, exendin-4, promoted bone regeneration mediated by the transplantation of adipose-derived stem cells in a metaphyseal defect mouse model of femur injury. However, the underlying mechanism is unclear. Bone imaging, immunohistochemistry real-time PCR and western blot analysis were used in the present study, and the results revealed that exendin-4 increased the transcription of the osteogenic differentiation-related genes and induced osteogenic differentiation in situ. Furthermore, the present data obtained from sorted adipose-derived stem cells revealed that exendin-4 promoted osteogenic differentiation and inhibited adipogenic differentiation in vitro. These findings indicated that exendin-4 facilitates osteogenic differentiation of transplanted adipose-derived stem cells for bone repair and illuminated clinical prospects of both adipose-derived stem cells and exendin-4 in stem-cell-based bone defect repair.
Collapse
Affiliation(s)
- Banglian Deng
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of Stomatology, Department of Oral Implantation, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Wenzhong Zhu
- Department of Stomatology, Shaanxi Province Geriatric Hospital, Xi'an, Shaanxi 710005, P.R. China
| | - Yansheng Duan
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of Stomatology, Department of Oral Implantation, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yuqian Hu
- Department of Stomatology, The Faculty of Medicine, Eastern University of Liaoning, Shenyang, Liaoning 110000, P.R. China
| | - Xuefeng Chen
- Xuefeng Dental Care Huaian, Huaian, Jiangsu 223000, P.R. China
| | - Shuang Song
- Health Science Center, Peking University, Beijing 100000, P.R. China
| | - Zian Yi
- Department of Stomatology, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Yingliang Song
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of Stomatology, Department of Oral Implantation, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
35
|
Srirussamee K, Mobini S, Cassidy NJ, Cartmell SH. Direct electrical stimulation enhances osteogenesis by inducing Bmp2 and Spp1 expressions from macrophages and preosteoblasts. Biotechnol Bioeng 2019; 116:3421-3432. [PMID: 31429922 PMCID: PMC6899728 DOI: 10.1002/bit.27142] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 08/03/2019] [Accepted: 08/09/2019] [Indexed: 12/16/2022]
Abstract
The capability of electrical stimulation (ES) in promoting bone regeneration has already been addressed in clinical studies. However, its mechanism is still being investigated and discussed. This study aims to investigate the responses of macrophages (J774A.1) and preosteoblasts (MC3T3-E1) to ES and the faradic by-products from ES. It is found that pH of the culture media was not significantly changed, whereas the average hydrogen peroxide concentration was increased by 3.6 and 5.4 µM after 1 and 2 hr of ES, respectively. The upregulation of Bmp2 and Spp1 messenger RNAs was observed after 3 days of stimulation, which is consistent among two cell types. It is also found that Spp1 expression of macrophages was partially enhanced by faradic by-products. Osteogenic differentiation of preosteoblasts was not observed during the early stage of ES as the level of Runx2 expression remains unchanged. However, cell proliferation was impaired by the excessive current density from the electrodes, and also faradic by-products in the case of macrophages. This study shows that macrophages could respond to ES and potentially contribute to the bone formation alongside preosteoblasts. The upregulation of Bmp2 and Spp1 expressions induced by ES could be one of the mechanisms behind the electrically stimulated osteogenesis.
Collapse
Affiliation(s)
| | - Sahba Mobini
- Instituto de Micro y Nanotecnología IMN-CNM, The Spanish National Research Council (CSIC), Madrid, Spain.,Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, Madrid, Spain
| | - Nigel J Cassidy
- Department of Civil Engineering, University of Birmingham, Birmingham, UK
| | - Sarah H Cartmell
- Department of Materials, The University of Manchester, Manchester, UK
| |
Collapse
|
36
|
Ma X, Fan C, Wang Y, Du Y, Zhu Y, Liu H, Lv L, Liu Y, Zhou Y. MiR-137 knockdown promotes the osteogenic differentiation of human adipose-derived stem cells via the LSD1/BMP2/SMAD4 signaling network. J Cell Physiol 2019; 235:909-919. [PMID: 31241766 DOI: 10.1002/jcp.29006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/06/2019] [Indexed: 12/18/2022]
Abstract
MicroRNAs are a group of endogenous regulators that participate in several cellular physiological processes. However, the role of miR-137 in the osteogenic differentiation of human adipose-derived stem cells (hASCs) has not been reported. This study verified a general downward trend in miR-137 expression during the osteogenic differentiation of hASCs. MiR-137 knockdown promoted the osteogenesis of hASCs in vitro and in vivo. Mechanistically, inhibition of miR-137 activated the bone morphogenetic protein 2 (BMP2)-mothers against the decapentaplegic homolog 4 (SMAD4) pathway, whereas repressed lysine-specific histone demethylase 1 (LSD1), which was confirmed as a negative regulator of osteogenesis in our previous studies. Furthermore, LSD1 knockdown enhanced the expression of BMP2 and SMAD4, suggesting the coordination of LSD1 in the osteogenic regulation of miR-137. This study indicated that miR-137 negatively regulated the osteogenic differentiation of hASCs via the LSD1/BMP2/SMAD4 signaling network, revealing a new potential therapeutic target of hASC-based bone tissue engineering.
Collapse
Affiliation(s)
- Xiaohan Ma
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Cong Fan
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China.,Department of General Dentistry II, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yuejun Wang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yangge Du
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yuan Zhu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Hao Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Longwei Lv
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, China.,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
37
|
Süloğlu AK, Karacaoğlu E, Bilgic HA, Selmanoğlu G, Koçkaya EA, Karaaslan C. Osteogenic differentiation of adipose tissue-derived mesenchymal stem cells on fibrin glue- or fibronectin-coated Ceraform®. J Biomater Appl 2019; 34:375-385. [PMID: 31165664 DOI: 10.1177/0885328219853421] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Aysun Kılıç Süloğlu
- 1 Hacettepe University, Faculty of Science, Department of Biology, Zoology Section, Beytepe Campus, Ankara, Turkey
| | - Elif Karacaoğlu
- 1 Hacettepe University, Faculty of Science, Department of Biology, Zoology Section, Beytepe Campus, Ankara, Turkey
| | - Hayriye Akel Bilgic
- 2 Hacettepe University, Faculty of Science, Department of Biology, Molecular Biology Section, Beytepe Campus, Ankara, Turkey
| | - Güldeniz Selmanoğlu
- 1 Hacettepe University, Faculty of Science, Department of Biology, Zoology Section, Beytepe Campus, Ankara, Turkey
| | - Evrim A Koçkaya
- 3 The Higher Vocational School of Health Services, Gazi University, Gölbaşı Campus, Ankara, Turkey
| | - Cagatay Karaaslan
- 2 Hacettepe University, Faculty of Science, Department of Biology, Molecular Biology Section, Beytepe Campus, Ankara, Turkey
| |
Collapse
|
38
|
Park MJ, Moon SJ, Baek JA, Lee EJ, Jung KA, Kim EK, Kim DS, Lee JH, Kwok SK, Min JK, Kim SJ, Park SH, Cho ML. Metformin Augments Anti-Inflammatory and Chondroprotective Properties of Mesenchymal Stem Cells in Experimental Osteoarthritis. THE JOURNAL OF IMMUNOLOGY 2019; 203:127-136. [PMID: 31142603 DOI: 10.4049/jimmunol.1800006] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/01/2019] [Indexed: 01/15/2023]
Abstract
Mesenchymal stem cells (MSCs) can protect against cartilage breakdown in osteoarthritis (OA) via their immunomodulatory capacities. However, the optimization strategy for using MSCs remains challenging. This study's objective was to identify the in vivo effects of metformin-stimulated adipose tissue-derived human MSCs (Ad-hMSCs) in OA. An animal model of OA was established by intra-articular injection of monosodium iodoacetate into rats. OA rats were divided into a control group and two therapy groups (treated with Ad-hMSCs or metformin-stimulated Ad-hMSCs). Limb nociception was assessed by measuring the paw withdrawal latency and threshold. Our data show that metformin increased IL-10 and IDO expression in Ad-hMSCs and decreased high-mobility group box 1 protein, IL-1β, and IL-6 expression. Metformin increased the migration capacity of Ad-hMSCs with upregulation of chemokine expression. In cocultures, metformin-stimulated Ad-hMSCs inhibited the mRNA expression of RUNX2, COL X, VEGF, MMP1, MMP3, and MMP13 in IL-1β-stimulated OA chondrocytes and increased the expression of TIMP1 and TIMP3. The antinociceptive activity and chondroprotective effects were greater in OA rats treated with metformin-stimulated Ad-hMSCs than in those treated with unstimulated Ad-hMSCs. TGF-β expression in subchondral bone of OA joints was attenuated more in OA rats treated with metformin-stimulated Ad-hMSCs. Our findings suggest that metformin offers a promising option for the clinical application of Ad-hMSCs as a cell therapy for OA.
Collapse
Affiliation(s)
- Min-Jung Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Su-Jin Moon
- Division of Rheumatology, Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Jin-Ah Baek
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Eun-Jung Lee
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Kyung-Ah Jung
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Eun-Kyung Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Da-Som Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Jung-Ho Lee
- Department of Plastic and Reconstructive Surgery, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - Seung-Ki Kwok
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Jun-Ki Min
- Division of Rheumatology, Department of Internal Medicine, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea; and
| | - Seok Jung Kim
- Department of Orthopedic Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea;
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul 137-701, South Korea;
| |
Collapse
|
39
|
Zhang J, Wu K, Xu T, Wu J, Li P, Wang H, Wu H, Wu G. Epigallocatechin-3-gallate enhances the osteoblastogenic differentiation of human adipose-derived stem cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:1311-1321. [PMID: 31114166 PMCID: PMC6485322 DOI: 10.2147/dddt.s192683] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Purpose The aim of this study is to investigate the effects of epigallocatechin-3-gallate (EGCG), a major polyphenol extracted from green tea, on the osteoblastogenic differentiation of human adipose-derived stem cells (hASCs). Patients and methods hASCs were acquired from human adipose tissue. With informed consent, subcutaneous adipose tissue samples were harvested from periorbital fat pad resections from ten healthy female adults who underwent double eyelid surgery. hASCs were cultured in osteogenic medium with or without EGCG (1 μM, 5 μM, or 10 μM) for 14 days. We evaluated the effects of EGCG by quantifying cell growth, ALP activity (an early osteoblastogenic differentiation marker), BSP, OCN (a late osteoblastogenic differentiation marker), and extracellular matrix mineralization. We also performed Western blots to measure osteoblastogenesis-related proteins such as Runx2 and adipoblastogenesis-related transcription factors, such as STAT3, C/EBP-α, and PPAR-γ. Results EGCG at 5 μM resulted in significantly higher cell proliferation and ALP activity than did the control on days 3, 7, and 14. On day 7, 5 μM EGCG significantly enhanced BSP expression. On day 14, EGCG at all concentrations promoted OCN expression. In addition, EGCG at 5 μM resulted in the highest level of extracellular matrix mineralization. On day 3, the expression levels of Runx2 were significantly higher in the 5 μM EGCG group than in the other groups, whereas later, on days 7 and 14, Runx2 expression levels in the EGCG group were significantly lower than those of the control group. EGCG at all three concentrations was associated with significantly lower levels of phosphorylated STAT3, C/EBP-α, and PPAR-γ. Conclusion EGCG at 5 μM significantly enhanced the osteoblastogenic differentiation of hASCs.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China,
| | - Kai Wu
- Spine Lab, Department of Orthopedic Surgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Ting Xu
- Department of Stomatology, First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Jiajun Wu
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China,
| | - Pengfei Li
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China,
| | - Hong Wang
- Department of Oral and Maxillofacial Surgery, Amsterdam University Medical Centre, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, North Holland, the Netherlands
| | - Huiling Wu
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China,
| | - Gang Wu
- Department of Oral Implantology and Prosthetic Dentistry, Academic Centre for Dentistry Amsterdam, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, North Holland, the Netherlands,
| |
Collapse
|
40
|
Trucillo E, Bisceglia B, Valdrè G, Giordano E, Reverchon E, Maffulli N, Della Porta G. Growth factor sustained delivery from poly-lactic-co-glycolic acid microcarriers and its mass transfer modeling by finite element in a dynamic and static three-dimensional environment bioengineered with stem cells. Biotechnol Bioeng 2019; 116:1777-1794. [PMID: 30905072 DOI: 10.1002/bit.26975] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/18/2019] [Accepted: 03/21/2019] [Indexed: 12/15/2022]
Abstract
Poly-lactic-co-glycolic acid (PLGA) microcarriers (0.8 ± 0.2 μm) have been fabricated with a load of 20 μg/gPLGA by an emulsion-based-proprietary technology to sustained deliver human bone morphogenetic protein 2 (hBMP2), a growth factor largely used for osteogenic induction. hBMP2 release profile, measured in vitro, showed a moderate "burst" release of 20% of the load in first 3 days, followed by a sustained release of 3% of the load along the following 21 days. PLGA microbeads loaded with fluorescent marker (8 mg/gPLGA ) and hydroxyapatite (30 mg/gPLGA ) were also fabricated and successfully dispersed within three-dimensional (3D) alginate scaffold (Ca-alginate 2% wt/wt) in a range between 50 and 200 mg/cm3 ; the presence of microcarriers within the scaffold induced a variation of its stiffness between 0.03 and 0.06 MPa; whereas the scaffold surface area was monitored always in the range of 190-200 m2 /g. Uniform microcarriers dispersion was obtained up to 200 mg/cm3 ; higher loading values in the 3D scaffold produced large aggregates. The release data and the surface area were, then, used to simulate by finite element modeling the hBMP2 mass transfer within the 3D hydrogel bioengineered with stem cells, in dynamic and static cultivations. The simulation was developed with COMSOL Multiphysics® giving a good representation of hBMP2 mass balances along microbeads (bulk eroded) and on cell surface (cell binding). hBMP2 degradation rate was also taken into account in the simulations. hBMP2 concentration of 20 ng/cm3 was set as a target because it has been described as the minimum effective value for stem cells stimulation versus the osteogenic phenotype. The sensitivity analysis suggested the best microbeads/cells ratio in the 3D microenvironment, along 21 days of cultivations in both static and dynamic cultivation (perfusion) conditions. The simulated formulation was so assembled experimentally using human mesenchymal stem cells and an improved scaffold stiffness up to 0.09 MPa (n = 3; p ≤ 0.01) was monitored after 21 days of cultivation; moreover a uniform extracellular matrix deposition within the 3D system was detected by Von Kossa staining, especially in dynamic conditions. The results indicated that the described tool can be useful for the design of 3D bioengineered microarchitecture by quantitative understanding.
Collapse
Affiliation(s)
- Emanuele Trucillo
- Department of Industrial Engineering, University of Salerno, Fisciano, SA, Italy
| | - Bruno Bisceglia
- Department of Industrial Engineering, University of Salerno, Fisciano, SA, Italy
| | - Giovanni Valdrè
- Department of Biology, Geology and Environmental Science, University of Bologna, Bologna, BO, Italy
| | - Emanuele Giordano
- Department of Electrical, Electronic and Information Engineering "Guglielmo Marconi" (DEI), University of Bologna, Cesena, FC, Italy
| | - Ernesto Reverchon
- Department of Industrial Engineering, University of Salerno, Fisciano, SA, Italy
| | - Nicola Maffulli
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, SA, Italy
| | - Giovanna Della Porta
- Department of Industrial Engineering, University of Salerno, Fisciano, SA, Italy.,Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, SA, Italy
| |
Collapse
|
41
|
Aziz AH, Bryant SJ. A comparison of human mesenchymal stem cell osteogenesis in poly(ethylene glycol) hydrogels as a function of MMP-sensitive crosslinker and crosslink density in chemically defined medium. Biotechnol Bioeng 2019; 116:1523-1536. [PMID: 30776309 DOI: 10.1002/bit.26957] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/28/2018] [Accepted: 02/14/2019] [Indexed: 12/13/2022]
Abstract
This study investigated osteogenesis of human mesenchymal stem cells encapsulated in matrix-metalloproteinase (MMP)-sensitive poly(ethylene glycol) (PEG) hydrogels in chemically defined medium (10 ng/ml bone morphogenic factor-2). Thiol-norbornene photoclick hydrogels were formed with CRGDS and crosslinkers of PEG dithiol (nondegradable), CVPLS-LYSGC (P1) or CRGRIGF-LRTDC (P2; dash indicates cleavage site) at two crosslink densities. Exogenous MMP-2 degraded P1 and P2 hydrogels similarly. MMP-14 degraded P1 hydrogels more rapidly than P2 hydrogels. Cell spreading was greatest in P1 low crosslinked hydrogels and to a lesser degree in P2 low crosslinked hydrogels, but not evident in nondegradable and high crosslinked MMP-sensitive hydrogels. Early osteogenesis (Alkaline phosphatase [ALP] activity) was accelerated in hydrogels that facilitated cell spreading. Contrarily, late osteogenesis (mineralization) was independent of cell spreading. Mineralized matrix was present in P1 hydrogels, but only present in P2 high crosslinked hydrogels and not yet present in nondegradable hydrogels. Overall, the low crosslinked P1 hydrogels exhibited an accelerated early and late osteogenesis with the highest ALP activity (Day 7), greatest calcium content (Day 14), and greatest collagen content (Day 28), concomitant with increased compressive modulus over time. Collectively, this study demonstrates that in chemically defined medium, hydrogel degradability is critical to accelerating early osteogenesis, but other factors are important in late osteogenesis.
Collapse
Affiliation(s)
- Aaron H Aziz
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado.,BioFrontiers Institute, University of Colorado, Boulder, Colorado
| | - Stephanie J Bryant
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado.,BioFrontiers Institute, University of Colorado, Boulder, Colorado.,Material Science and Engineering, University of Colorado, Boulder, Colorado
| |
Collapse
|
42
|
Oberoi G, Nitsch S, Edelmayer M, Janjić K, Müller AS, Agis H. 3D Printing-Encompassing the Facets of Dentistry. Front Bioeng Biotechnol 2018; 6:172. [PMID: 30525032 PMCID: PMC6262086 DOI: 10.3389/fbioe.2018.00172] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/29/2018] [Indexed: 12/14/2022] Open
Abstract
This narrative review presents an overview on the currently available 3D printing technologies and their utilization in experimental, clinical and educational facets, from the perspective of different specialties of dentistry, including oral and maxillofacial surgery, orthodontics, endodontics, prosthodontics, and periodontics. It covers research and innovation, treatment modalities, education and training, employing the rapidly developing 3D printing process. Research-oriented advancement in 3D printing in dentistry is witnessed by the rising number of publications on this topic. Visualization of treatment outcomes makes it a promising clinical tool. Educational programs utilizing 3D-printed models stimulate training of dental skills in students and trainees. 3D printing has enormous potential to ameliorate oral health care in research, clinical treatment, and education in dentistry.
Collapse
Affiliation(s)
- Gunpreet Oberoi
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Center for Medical Physics and Biomedical Engineering, Medical University Vienna, Vienna, Austria
| | - Sophie Nitsch
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria.,Department of Health Sciences, FH Wien, University of Applied Sciences, Vienna, Austria
| | - Michael Edelmayer
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Department of Oral Surgery, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Klara Janjić
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Anna Sonja Müller
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Hermann Agis
- Department of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
43
|
Lin X, Hunziker EB, Liu T, Hu Q, Liu Y. Enhanced biocompatibility and improved osteogenesis of coralline hydroxyapatite modified by bone morphogenetic protein 2 incorporated into a biomimetic coating. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 96:329-336. [PMID: 30606540 DOI: 10.1016/j.msec.2018.11.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 08/31/2018] [Accepted: 11/13/2018] [Indexed: 11/17/2022]
Abstract
OBJECTIVES (1) To determine whether the biocompatibility of coralline hydroxyapatite (CHA) granules could be improved by using an octacalcium phosphate (OCP) coating layer, and/or functionalized with bone morphogenetic protein 2 (BMP-2), and (2) to investigate if BMP-2 incorporated into this coating is able to enhance its osteoinductive efficiency, in comparison to its surface-adsorbed delivery mode. METHODS CHA granules (0.25 g per sample) bearing a coating-incorporated depot of BMP-2 (20 μg/sample) together with the controls (CHA bearing an adsorbed depot of BMP-2; CHA granules with an OCP coating without BMP-2; pure CHA granules) were implanted subcutaneously in rats (n = 6 animals per group). Five weeks later, the implants were retrieved for histomorphometric analysis to quantify the volume of newly generated bone, bone marrow, fibrous tissue and foreign body giant cells (FBGCs). The osteoinductive efficiency of BMP-2 and the rates of CHA degradation were also determined. RESULTS The group with an OCP coating-incorporated depot of BMP-2 showed the highest volume and quality or bone, and the highest osteoinductive efficacy. OCP coating was able to reduce inflammatory responses (improve biocompatibility), and also simple adsorption of BMP-2 to CHA achieved this. CONCLUSIONS The biocompatibility of CHA granules (reduction of inflammation) was significantly improved by coating with a layer of OCP. Pure surface adsorption of BMP-2 to CHA also reduced inflammation. Incorporation of BMP-2 into the OCP coatings was associated with the highest volume and quality of bone, and the highest biocompatibility degree of the CHA granules. CLINICAL SIGNIFICANCE Higher osteoinductivity and improved biocompatibility of CHA can be obtained when a layer of BMP-2 functionalized OCP is deposited on the surfaces of CHA granules.
Collapse
Affiliation(s)
- Xingnan Lin
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, 210008 Nanjing, China; Department of Oral Implantology and Prosthetic Dentistry, Academic Centre of Dentistry Amsterdam (ACTA), VU University and University of Amsterdam, 1081LA Amsterdam, the Netherlands.
| | - Ernst B Hunziker
- Departments of Osteoporosis and Orthopaedic Surgery, Inselspital (University Hospital), Bern, 3010 Bern, Switzerland.
| | - Tie Liu
- Department of Oral Implantology, Hospital/School of Stomatology, Zhejiang University, 310003 Hangzhou, Zhejiang, China
| | - Qingang Hu
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Medical School, Nanjing University, 210008 Nanjing, China.
| | - Yuelian Liu
- Department of Oral Implantology and Prosthetic Dentistry, Academic Centre of Dentistry Amsterdam (ACTA), VU University and University of Amsterdam, 1081LA Amsterdam, the Netherlands.
| |
Collapse
|
44
|
Graceffa V, Vinatier C, Guicheux J, Stoddart M, Alini M, Zeugolis DI. Chasing Chimeras - The elusive stable chondrogenic phenotype. Biomaterials 2018; 192:199-225. [PMID: 30453216 DOI: 10.1016/j.biomaterials.2018.11.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/02/2018] [Accepted: 11/09/2018] [Indexed: 12/27/2022]
Abstract
The choice of the best-suited cell population for the regeneration of damaged or diseased cartilage depends on the effectiveness of culture conditions (e.g. media supplements, three-dimensional scaffolds, mechanical stimulation, oxygen tension, co-culture systems) to induce stable chondrogenic phenotype. Herein, advances and shortfalls in in vitro, preclinical and clinical setting of various in vitro microenvironment modulators on maintaining chondrocyte phenotype or directing stem cells towards chondrogenic lineage are critically discussed. Chondrocytes possess low isolation efficiency, limited proliferative potential and rapid phenotypic drift in culture. Mesenchymal stem cells are relatively readily available, possess high proliferation potential, exhibit great chondrogenic differentiation capacity, but they tend to acquire a hypertrophic phenotype when exposed to chondrogenic stimuli. Embryonic and induced pluripotent stem cells, despite their promising in vitro and preclinical data, are still under-investigated. Although a stable chondrogenic phenotype remains elusive, recent advances in in vitro microenvironment modulators are likely to develop clinically- and commercially-relevant therapies in the years to come.
Collapse
Affiliation(s)
- Valeria Graceffa
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Claire Vinatier
- INSERMU1229, Regenerative Medicine and Skeleton (RMeS), University of Nantes, UFR Odontologie & CHU Nantes, PHU 4 OTONN, 44042 Nantes, France
| | - Jerome Guicheux
- INSERMU1229, Regenerative Medicine and Skeleton (RMeS), University of Nantes, UFR Odontologie & CHU Nantes, PHU 4 OTONN, 44042 Nantes, France
| | - Martin Stoddart
- AO Research Institute, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Mauro Alini
- AO Research Institute, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland.
| |
Collapse
|
45
|
Tsai YT, Huang CW, Liu HY, Huang MC, Sun TP, Chen WC, Wu CY, Ding ST, Chen HY. Enhanced bone morphogenic property of parylene-C. Biomater Sci 2018; 4:1754-1760. [PMID: 27782270 DOI: 10.1039/c6bm00664g] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The ability to induce osteointegration was introduced to a parylene-C surface via the simple and intuitive process of protein adsorption mediated by hydrophobic interactions. In this way, bone morphogenetic protein (BMP)-2, fibronectin, and platelet-rich plasma (PRP) could be immobilized on parylene-C surfaces. This approach alleviates concerns related to the use of potentially harmful substances in parylene-C modification processes. The adsorbed protein molecules were quantitatively characterized with respect to adsorption efficacy and binding affinity, and the important biological activities of the proteins were also examined using both early and late markers of osteogenetic activity, including alkaline phosphatase expression, calcium mineralization and marker gene expression. Additionally, the adsorbed PRP exhibited potential as a substitute for expensive recombinant growth factors by effectively inducing comparable osteogenetic activity. In addition to the excellent biocompatibility of parylene-C and its ability to coat a wide variety of substrate materials, the modification of parylene-C via protein adsorption provides unlimited possibilities for installing specific biological functions, expanding the potential applications of this material to include various biointerface platforms.
Collapse
Affiliation(s)
- Ya-Ting Tsai
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan.
| | - Chao-Wei Huang
- Department of Animal Science and Technology, National Taiwan University, Taipei 10617, Taiwan.
| | - Hui-Yu Liu
- Department of Animal Science and Technology, National Taiwan University, Taipei 10617, Taiwan.
| | - Mei-Ching Huang
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan.
| | - Ting-Pi Sun
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan.
| | - Wen-Chien Chen
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, College of Medicine Chang Gung University, Taoyuan 333, Taiwan.
| | - Chih-Yu Wu
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan.
| | - Shih-Torng Ding
- Department of Animal Science and Technology, National Taiwan University, Taipei 10617, Taiwan.
| | - Hsien-Yeh Chen
- Department of Chemical Engineering, National Taiwan University, Taipei 10617, Taiwan.
| |
Collapse
|
46
|
Ort C, Dayekh K, Xing M, Mequanint K. Emerging Strategies for Stem Cell Lineage Commitment in Tissue Engineering and Regenerative Medicine. ACS Biomater Sci Eng 2018; 4:3644-3657. [PMID: 33429592 DOI: 10.1021/acsbiomaterials.8b00532] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Stem cells have transformed the fields of tissue engineering and regenerative medicine, and their potential to further advance these fields cannot be overstated. The stem cell niche is a dynamic microenvironment that determines cell fate during development and tissue repair following an injury. Classically, stem cells were studied in isolation of their microenvironment; however, contemporary research has produced a myriad of evidence that shows the importance of multiple aspects of the stem cell niche in regulating their processes. In the context of tissue engineering and regenerative medicine studies, the niche is an artificial environment provided by culture conditions. In vitro culture conditions may involve coculturing with other cell types, developing specific biomaterials, and applying relevant forces to promote the desired lineage commitment. Considerable advance has been made over the past few years toward directed stem cell differentiation; however, the unspecific differentiation of stem cells yielding a mixed population of cells has been a challenge. In this review, we provide a systematic review of the emerging strategies used for lineage commitment within the context of tissue engineering and regenerative medicine. These strategies include scaffold pore-size and pore-shape gradients, stress relaxation, sonic and electromagnetic effects, and magnetic forces. Finally, we provide insights and perspectives into future directions focusing on signaling pathways activated during lineage commitment using external stimuli.
Collapse
Affiliation(s)
| | | | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, 66 Chancellors Circle, Winnipeg R3T 2N2, Canada
| | | |
Collapse
|
47
|
Dubey NK, Mishra VK, Dubey R, Deng YH, Tsai FC, Deng WP. Revisiting the Advances in Isolation, Characterization and Secretome of Adipose-Derived Stromal/Stem Cells. Int J Mol Sci 2018; 19:ijms19082200. [PMID: 30060511 PMCID: PMC6121360 DOI: 10.3390/ijms19082200] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/08/2018] [Accepted: 07/24/2018] [Indexed: 12/13/2022] Open
Abstract
Adipose-derived stromal/stem cells (ASCs) seems to be a promising regenerative therapeutic agent due to the minimally invasive approach of their harvest and multi-lineage differentiation potential. The harvested adipose tissues are further digested to extract stromal vascular fraction (SVF), which is cultured, and the anchorage-dependent cells are isolated in order to characterize their stemness, surface markers, and multi-differentiation potential. The differentiation potential of ASCs is directed through manipulating culture medium composition with an introduction of growth factors to obtain the desired cell type. ASCs have been widely studied for its regenerative therapeutic solution to neurologic, skin, wound, muscle, bone, and other disorders. These therapeutic outcomes of ASCs are achieved possibly via autocrine and paracrine effects of their secretome comprising of cytokines, extracellular proteins and RNAs. Therefore, secretome-derivatives might offer huge advantages over cells through their synthesis and storage for long-term use. When considering the therapeutic significance and future prospects of ASCs, this review summarizes the recent developments made in harvesting, isolation, and characterization. Furthermore, this article also provides a deeper insight into secretome of ASCs mediating regenerative efficacy.
Collapse
Affiliation(s)
- Navneet Kumar Dubey
- Ceramics and Biomaterials Research Group, Advanced Institute of Materials Science, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam.
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam.
| | - Viraj Krishna Mishra
- Applied Biotech Engineering Centre (ABEC), Department of Biotechnology, Ambala College of Engineering and Applied Research, Ambala 133101, India.
| | - Rajni Dubey
- Graduate Institute Food Science and Technology, National Taiwan University, Taipei 10617, Taiwan.
| | - Yue-Hua Deng
- Stem Cell Research Center, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Life Science, Fu Jen Catholic University, New Taipei City 24205, Taiwan.
| | - Feng-Chou Tsai
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Win-Ping Deng
- Stem Cell Research Center, Taipei Medical University, Taipei 11031, Taiwan.
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Basic medicine, Fu-Jen Catholic University, New Taipei City 24205, Taiwan.
| |
Collapse
|
48
|
Su W, Ma X, Sun Z, Yi Z, Cui X, Chen G, Chen X, Guo B, Li X. RhBMP-2 and concomitant rapid material degradation synergistically promote bone repair and regeneration with collagen-hydroxyapatite nanocomposites. J Mater Chem B 2018; 6:4338-4350. [PMID: 32254509 DOI: 10.1039/c8tb00405f] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The effective treatment of bone defects is still a great challenge in clinical practice. Synthetic bone-grafting substitutes of composition and structure analogous to bone as well as incorporated with growth factors are considered to be a promising solution. In this study, a collagen-hydroxyapatite (CHA) nanocomposite scaffold was developed by collagen self-assembly with simultaneous HA synthesis. The physicochemical properties such as morphology, inorganic phase, thermal decomposition, specific surface area and pore size distribution were characterized. The osteogenicity of CHA in the absence or presence of recombinant human bone morphogenetic protein-2 (rhBMP-2) was assessed both by cell culturing and animal implantation experiments. The gene expression results showed that the osteogenic differentiation capacity of rat bone mesenchymal stem cells (rBMSCs) has been enhanced both by CHA and rhBMP-2. The efficient bone regeneration of femoral defects in rabbits was achieved with CHA and CHA pre-absorbed rhBMP-2 (CHA/B), confirmed by micro-computed tomography measurements, histological observation and immunohistochemical analyses. The CHA nanocomposite was completely degraded within 8 weeks and replaced by new bone. It was found that rhBMP-2 not only accelerated and enhanced bone formation, but also expedited the degradation of CHA. It is believed that the rhBMP-2 and concomitant rapid material degradation synergistically promote bone repair and regeneration with CHA. The biodegradation behavior of CHA in the presence of rhBMP-2 can be further investigated to gain an in-depth understanding of the complex interplays among biomaterials, growth factors and their target cells. The relevant knowledge will facilitate the search for a reasonable, safe and efficient methodology for the introduction of growth factors to biomaterials so as to achieve satisfactory tissue regeneration.
Collapse
Affiliation(s)
- Wen Su
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, Sichuan, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Zhang S, Ma B, Liu F, Duan J, Wang S, Qiu J, Li D, Sang Y, Liu C, Liu D, Liu H. Polylactic Acid Nanopillar Array-Driven Osteogenic Differentiation of Human Adipose-Derived Stem Cells Determined by Pillar Diameter. NANO LETTERS 2018. [PMID: 29517915 DOI: 10.1021/acs.nanolett.7b04747] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Numerous studies have determined that physical cues, especially the nanotopography of materials, play key roles in directing stem cell differentiation. However, most research on nanoarrays for stem cell fate regulation is based on nonbiodegradable materials, such as silicon wafers, TiO2, and poly(methyl methacrylate), which are rarely used as tissue engineering biomaterials. In this study, we prepared biodegradable polylactic acid (PLA) nanopillar arrays with different diameters but the same center-to-center distance using a series of anodic aluminum oxide nanowell arrays as templates. Human adipose-derived stem cells (hADSCs) were selected to investigate the effect of the diameter of PLA nanopillar arrays on stem cell differentiation. By culturing hADSCs without the assistance of any growth factors or osteogenic-induced media, the differentiation tendencies of hADSCs on the nanopillar arrays were assessed at the gene and protein levels. The assessment results suggested that the osteogenic differentiation of hADSCs can be driven by nanopillar arrays, especially by nanopillar arrays with a diameter of 200 nm. Moreover, an in vivo animal model of the samples demonstrated that PLA film with the 200 nm pillar array exhibits an improved ectopic osteogenic ability compared with the planar PLA film after 4 weeks of ectopic implantation. This study has provided a new variable to investigate in the interaction between stem cells and nanoarray structures, which will guide the bone regeneration clinical research field. This work paves the way for the utility of degradable biopolymer nanoarrays with specific geometrical and mechanical signals in biomedical applications, such as patches and strips for spine fusion, bone crack repair, and restoration of tooth enamel.
Collapse
Affiliation(s)
- Shan Zhang
- State Key Laboratory of Crystal Materials , Shandong University , Jinan , 250100 , China
| | - Baojin Ma
- State Key Laboratory of Crystal Materials , Shandong University , Jinan , 250100 , China
| | - Feng Liu
- State Key Laboratory of Crystal Materials , Shandong University , Jinan , 250100 , China
| | - Jiazhi Duan
- State Key Laboratory of Crystal Materials , Shandong University , Jinan , 250100 , China
| | - Shicai Wang
- State Key Laboratory of Crystal Materials , Shandong University , Jinan , 250100 , China
| | - Jichuan Qiu
- State Key Laboratory of Crystal Materials , Shandong University , Jinan , 250100 , China
| | - Dong Li
- Cryomedicine Laboratory , Qilu Hospital, Shandong University , Jinan , 250012 , China
| | - Yuanhua Sang
- State Key Laboratory of Crystal Materials , Shandong University , Jinan , 250100 , China
| | - Chao Liu
- Department of Oral and Maxillofacial surgery, Qilu Hospital, Institute of Stomatology , Shandong University , Jinan , 250012 , China
| | - Duo Liu
- State Key Laboratory of Crystal Materials , Shandong University , Jinan , 250100 , China
| | - Hong Liu
- State Key Laboratory of Crystal Materials , Shandong University , Jinan , 250100 , China
- Institute for Advanced Interdisciplinary Research , Jinan University , Jinan , 250022 , China
| |
Collapse
|
50
|
Palade J, Djordjevic D, Hutchins ED, George RM, Cornelius JA, Rawls A, Ho JWK, Kusumi K, Wilson-Rawls J. Identification of satellite cells from anole lizard skeletal muscle and demonstration of expanded musculoskeletal potential. Dev Biol 2018; 433:344-356. [PMID: 29291980 PMCID: PMC6180209 DOI: 10.1016/j.ydbio.2017.08.037] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 08/22/2017] [Accepted: 08/29/2017] [Indexed: 10/18/2022]
Abstract
The lizards are evolutionarily the closest vertebrates to humans that demonstrate the ability to regenerate entire appendages containing cartilage, muscle, skin, and nervous tissue. We previously isolated PAX7-positive cells from muscle of the green anole lizard, Anolis carolinensis, that can differentiate into multinucleated myotubes and express the muscle structural protein, myosin heavy chain. Studying gene expression in these satellite/progenitor cell populations from A. carolinensis can provide insight into the mechanisms regulating tissue regeneration. We generated a transcriptome from proliferating lizard myoprogenitor cells and compared them to transcriptomes from the mouse and human tissues from the ENCODE project using XGSA, a statistical method for cross-species gene set analysis. These analyses determined that the lizard progenitor cell transcriptome was most similar to mammalian satellite cells. Further examination of specific GO categories of genes demonstrated that among genes with the highest level of expression in lizard satellite cells were an increased number of genetic regulators of chondrogenesis, as compared to mouse satellite cells. In micromass culture, lizard PAX7-positive cells formed Alcian blue and collagen 2a1 positive nodules, without the addition of exogenous morphogens, unlike their mouse counterparts. Subsequent quantitative RT-PCR confirmed up-regulation of expression of chondrogenic regulatory genes in lizard cells, including bmp2, sox9, runx2, and cartilage specific structural genes, aggrecan and collagen 2a1. Taken together, these data suggest that tail regeneration in lizards involves significant alterations in gene regulation with expanded musculoskeletal potency.
Collapse
Affiliation(s)
- Joanna Palade
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, USA.
| | - Djordje Djordjevic
- Bioinformatics and Systems Medicine Laboratory, Victor Chang Cardiac Research Institute and St. Vincent's Clinical School, The University of New South Wales, Darlinghurst, NSW 2010, Australia.
| | - Elizabeth D Hutchins
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, USA; Neurogenomics Division, Translational Genomics Research Institute, 455 N. Fifth Street Phoenix, 85004, AZ, USA.
| | - Rajani M George
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, USA.
| | - John A Cornelius
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, USA.
| | - Alan Rawls
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, USA.
| | - Joshua W K Ho
- Bioinformatics and Systems Medicine Laboratory, Victor Chang Cardiac Research Institute and St. Vincent's Clinical School, The University of New South Wales, Darlinghurst, NSW 2010, Australia.
| | - Kenro Kusumi
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, USA; Neurogenomics Division, Translational Genomics Research Institute, 455 N. Fifth Street Phoenix, 85004, AZ, USA.
| | - Jeanne Wilson-Rawls
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, USA.
| |
Collapse
|