1
|
Khan NM, Wilderman A, Kaiser JM, Kamalakar A, Goudy SL, Cotney J, Drissi H. Enhanced osteogenic potential of iPSC-derived mesenchymal progenitor cells following genome editing of GWAS variants in the RUNX1 gene. Bone Res 2024; 12:70. [PMID: 39643619 PMCID: PMC11624199 DOI: 10.1038/s41413-024-00369-x] [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: 03/19/2024] [Revised: 08/19/2024] [Accepted: 09/03/2024] [Indexed: 12/09/2024] Open
Abstract
Recent genome-wide association studies (GWAS) identified 518 significant loci associated with bone mineral density (BMD), including variants at the RUNX1 locus (rs13046645, rs2834676, and rs2834694). However, their regulatory impact on RUNX1 expression and bone formation remained unclear. This study utilized human induced pluripotent stem cells (iPSCs) differentiated into osteoblasts to investigate these variants' regulatory roles. CRISPR/Cas9 was employed to generate mutant (Δ) iPSC lines lacking these loci at the RUNX1 locus. Deletion lines (Δ1 and Δ2) were created in iPSCs to assess the effects of removing regions containing these loci. Deletion lines exhibited enhanced osteogenic potential, with increased expression of osteogenic marker genes and Alizarin Red staining. Circularized chromosome conformation capture (4C-Seq) was utilized to analyze interactions between BMD-associated loci and the RUNX1 promoter during osteogenesis. Analysis revealed altered chromatin interactions with multiple gene promoters including RUNX1 isoform, as well as SETD4, a histone methyltransferase, indicating their regulatory influence. Interestingly, both deletion lines notably stimulated the expression of the long isoform of RUNX1, with more modest effects on the shorter isoform. Consistent upregulation of SETD4 and other predicted targets within the Δ2 deletion suggested its removal removed a regulatory hub constraining expression of multiple genes at this locus. In vivo experiments using a bone defect model in mice demonstrated increased bone regeneration with homozygous deletion of the Δ2 region. These findings indicate that BMD-associated variants within the RUNX1 locus regulate multiple effector genes involved in osteoblast commitment, providing valuable insights into genetic regulation of bone density and potential therapeutic targets.
Collapse
Affiliation(s)
- Nazir M Khan
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Decatur, GA, USA
| | - Andrea Wilderman
- Department of Genetics and Genome Sciences, University of Connecticut, Farmington, CT, USA
| | - Jarred M Kaiser
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Decatur, GA, USA
| | - Archana Kamalakar
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA, USA
| | - Steven L Goudy
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA, USA
| | - Justin Cotney
- Department of Genetics and Genome Sciences, University of Connecticut, Farmington, CT, USA
| | - Hicham Drissi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA.
- Atlanta VA Medical Center, Decatur, GA, USA.
| |
Collapse
|
2
|
Krukiewicz K, Contessotto P, Nedjari S, Martino MM, Redenski I, Gabet Y, Speranza G, O'Brien T, Altankov G, Awaja F. Clinical potential of plasma-functionalized graphene oxide ultrathin sheets for bone and blood vessel regeneration: Insights from cellular and animal models. BIOMATERIALS ADVANCES 2024; 161:213867. [PMID: 38669824 DOI: 10.1016/j.bioadv.2024.213867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 04/01/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024]
Abstract
Graphene and graphene oxide (GO), due to their unique chemical and physical properties, possess biochemical characteristics that can trigger intercellular signals promoting tissue regeneration. Clinical applications of thin GO-derived sheets have inspired the development of various tissue regeneration and repair approaches. In this study, we demonstrate that ultrathin sheets of plasma-functionalized and reduced GO, with the oxygen content ranging from 3.2 % to 22 % and the nitrogen content from 0 % to 8.3 %, retain their essential mechanical and molecular integrity, and exhibit robust potential for regenerating bone tissue and blood vessels across multiple cellular and animal models. Initially, we observed the growth of blood vessels and bone tissue in vitro using these functionalized GO sheets on human adipose-derived mesenchymal stem cells and umbilical vein endothelial cells. Remarkably, our study indicates a 2.5-fold increase in mineralization and two-fold increase in tubule formation even in media lacking osteogenic and angiogenic supplements. Subsequently, we observed the initiation, conduction, and formation of bone and blood vessels in a rat tibial osteotomy model, evident from a marked 4-fold increase in the volume of low radio-opacity bone tissue and a significant elevation in connectivity density, all without the use of stem cells or growth factors. Finally, we validated these findings in a mouse critical-size calvarial defect model (33 % higher healing rate) and a rat skin lesion model (up to 2.5-fold increase in the number of blood vessels, and 35 % increase in blood vessels diameter). This study elucidates the pro-osteogenic and pro-angiogenic properties of both pristine and plasma-treated GO ultrathin films. These properties suggest their significant potential for clinical applications, and as valuable biomaterials for investigating fundamental aspects of bone and blood vessel regeneration.
Collapse
Affiliation(s)
- Katarzyna Krukiewicz
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, Gliwice, Poland; Centre for Organic and Nanohybrid Electronics, Silesian University of Technology, Gliwice, Poland.
| | - Paolo Contessotto
- Department of Molecular Medicine, Università degli Studi di Padova, Padua, Italy.
| | - Salima Nedjari
- Molecular Dynamics at Cell-Biomaterial Interface, Institute for Bioengineering of Catalonia, Barcelona, Spain
| | - Mikaël M Martino
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia.
| | - Idan Redenski
- Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Yankel Gabet
- Department of Anatomy and Anthropology, Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel.
| | | | - Timothy O'Brien
- Centre for Research in Medical Devices, University of Galway, Galway, Ireland.
| | - George Altankov
- ICREA & Institute for Bioengineering of Catalonia, Barcelona, Spain; Medical University Pleven, Bulgaria
| | - Firas Awaja
- Department of Medicine, University of Galway, Galway, Ireland; Engmat Ltd., Clybaun Road, Galway, Ireland.
| |
Collapse
|
3
|
Zhuikova YV, Zhuikov VA, Makhina TK, Efremov YM, Aksenova NA, Timashev PS, Bonartseva GA, Varlamov VP. Preparation and characterization of poly(3-hydroxybutyrate)/chitosan composite films using acetic acid as a solvent. Int J Biol Macromol 2023; 248:125970. [PMID: 37494998 DOI: 10.1016/j.ijbiomac.2023.125970] [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: 05/19/2023] [Revised: 06/27/2023] [Accepted: 07/22/2023] [Indexed: 07/28/2023]
Abstract
Poly(3-hydroxybutyrate) and chitosan are among the most widely used polymers for biomedical applications due to their biocompatibility, renewability and low toxicity. The creation of composite materials based on biopolymers belonging to different classes makes it possible to overcome the disadvantages of each of the components and to obtain a material with specific properties. Solving this problem is associated with difficulties in the selection of conditions and solvents for obtaining the composite material. In our study, acetic acid was used as a common solvent for hydrophobic poly(3-hydroxybutyrate) and chitosan. Mechanical, thermal, physicochemical and surface properties of the composites and homopolymers were investigated. The composite films had less crystallinity and hydrophobicity than poly(3-hydroxybutyrate), and the addition of chitosan caused an increase in moisture absorption, a decrease in contact angle and changes in mechanical properties of the poly(3-hydroxybutyrate). The inclusion of varying amounts of chitosan controlled the properties of the composite, which will be important in the future for its specific biomedical applications.
Collapse
Affiliation(s)
- Yulia V Zhuikova
- Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russia.
| | - Vsevolod A Zhuikov
- Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russia
| | - Tatiana K Makhina
- Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russia
| | - Yuri M Efremov
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Nadezhda A Aksenova
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia; N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - Peter S Timashev
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia; World-Class Research Center "Digital Biodesign and Personalized Healthcare" Moscow, Russia; Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| | - Garina A Bonartseva
- Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russia
| | - Valery P Varlamov
- Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
4
|
Conrad B, Yang F. Hydroxyapatite-coated gelatin microribbon scaffolds induce rapid endogenous cranial bone regeneration in vivo. BIOMATERIALS ADVANCES 2022; 140:213050. [PMID: 35917686 DOI: 10.1016/j.bioadv.2022.213050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/25/2022] [Accepted: 07/24/2022] [Indexed: 06/15/2023]
Abstract
Hydroxyapatite (HA) has a composition similar to mineral bone and has been used for coating macroporous scaffolds to enhance bone formation. However, previous macroporous scaffolds did not support minimally invasive delivery. Our lab has reported on gelatin-based microribbon (μRB) shaped hydrogels, which combine injectability with macroporosity and support cranial bone formation in an immunocompromised mouse model. However, gelatin alone was not sufficient to support cranial bone formation in immunocompetent animals. To overcome this challenge, here we evaluated two methods to incorporate HA into gelatin μRB scaffolds using either modified simulated body fluid (mSBF) or commercially available HA nanoparticles (HAnp). HA incorporation and distribution were characterized using scanning electron microscopy and energy-dispersive X-ray spectroscopy. While both methods enhanced MSC osteogenesis and mineralization, the mSBF method led to undesirable reduction in mechanical properties. HAnp-coated μRB scaffolds were further evaluated in an immunocompetent mouse cranial defect model. Acellular HAnp-coated gelatin μRB scaffolds induced rapid and robust endogenous cranial bone regeneration as shown by MicroCT imaging and histology. Co-delivery with exogenous MSCs led to later bone resorption accompanied by increased osteoclast activity. In summary, our results demonstrate the promise of gelatin μRBs with HAnps as a promising therapy for cranial bone regeneration without the need for exogenous cells or growth factors.
Collapse
Affiliation(s)
- Bogdan Conrad
- Program of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 240 Pasteur Dr., Biomedical Innovation Building 1200, Palo Alto, CA 94304, United States of America.
| | - Fan Yang
- Departments of Orthopaedic Surgery and Bioengineering Stanford University, 240 Pasteur Dr., Biomedical Innovation Building 1200, Palo Alto, CA 94304, United States of America.
| |
Collapse
|
5
|
Shibli JA, Nagay BE, Suárez LJ, Urdániga Hung C, Bertolini M, Barão VAR, Souza JGS. Bone Tissue Engineering Using Osteogenic Cells: From the Bench to the Clinical Application. Tissue Eng Part C Methods 2022; 28:179-192. [PMID: 35166162 DOI: 10.1089/ten.tec.2022.0021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The use of tissue engineering to restore and to build new bone tissue is under active research at present. The following review summarizes the latest studies and clinical trials related to the use of osteogenic cells, biomaterials, and scaffolds to regenerate bone defects in the human jaws. Bone tissue engineering (BTE) combined with scaffolds have provided a range of advantages not only to transport the target cells to their desired destination but also to support the early phases of the mineralization process. The mechanical, chemical, and physical properties of scaffolds have been evaluated as they affect the quantity of bone regeneration, particularly in the oral cavity. This review also highlighted the mechanisms underlying bone homeostasis, including the key transcription factors and signaling pathways responsible for regulating the differentiation of osteoblast lineage. Furthering understanding of the mechanisms of cellular signaling in skeletal remodeling with the use of mesenchymal stem cells and the proper scaffold properties are key-factors to enable the incorporation of new and effective treatment methods into clinical practice for bone tissue regeneration using BTE. Impact Statement The use of mesenchymal stem cells able to differentiate in osteoblast lineage for bone tissue engineering (BTE) remains a major challenge. Viable cells and signaling pathways play an essential role in bone repair and regeneration of critical size defects. Recent advances in scaffolds and biological factors such as growth factors (e.g., cytokines and hormones) controlling the osteogenic signaling cascade are now becoming new players affecting the osteogenic potential of cells. Such techniques will significantly impact the maxillofacial bone tissue replacement, repair, and regeneration for patients without having to rely on donor banks or other surgical sites.
Collapse
Affiliation(s)
- Jamil Awad Shibli
- Dental Research Division, Department of Periodontology, Guarulhos University, Praça Tereza Cristina, Guarulhos, Brazil
| | - Bruna Egumi Nagay
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Brazil
| | - Lina J Suárez
- Dental Research Division, Department of Periodontology, Guarulhos University, Praça Tereza Cristina, Guarulhos, Brazil.,Departamento de Ciencias Básicas y Medicina Oral, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Celeste Urdániga Hung
- Dental Research Division, Department of Periodontology, Guarulhos University, Praça Tereza Cristina, Guarulhos, Brazil
| | - Martinna Bertolini
- Department of Periodontics and Preventive Dentistry, University of Pittsburgh School of Dental Medicine, Pittsburgh, Pennsylvania, USA
| | - Valentim A R Barão
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Brazil
| | - João Gabriel S Souza
- Dental Research Division, Department of Periodontology, Guarulhos University, Praça Tereza Cristina, Guarulhos, Brazil.,Dental Science School (Faculdade de Ciências Odontológicas-FCO), Montes Claros, Brazil
| |
Collapse
|
6
|
Bonartsev A, Voinova V, Volkov A, Muraev A, Boyko E, Venediktov A, Didenko N, Dolgalev A. Scaffolds Based on Poly(3-Hydroxybutyrate) and Its Copolymers for Bone Tissue Engineering (Review). Sovrem Tekhnologii Med 2022; 14:78-90. [PMID: 37181830 PMCID: PMC10171059 DOI: 10.17691/stm2022.14.5.07] [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: 03/02/2022] [Indexed: 05/16/2023] Open
Abstract
Biodegradable and biocompatible polymers are actively used in tissue engineering to manufacture scaffolds. Biomedical properties of polymer scaffolds depend on the physical and chemical characteristics and biodegradation kinetics of the polymer material, 3D microstructure and topography of the scaffold surface, as well as availability of minerals, medicinal agents, and growth factors loaded into the scaffold. However, in addition to the above, the intrinsic biological activity of the polymer and its biodegradation products can also become evident. This review provides studies demonstrating that scaffolds made of poly(3-hydroxybutyrate) (PHB) and its copolymers have their own biological activity, and namely, osteoinductive properties. PHB can induce differentiation of mesenchymal stem cells in the osteogenic direction in vitro and stimulates bone tissue regeneration during the simulation of critical and non-critical bone defects in vivo.
Collapse
Affiliation(s)
- A.P. Bonartsev
- Associate Professor, Department of Bioengineering, Faculty of Biology; Lomonosov Moscow State University, 1–12 Leninskiye Gory, Moscow, 119234, Russia
- Corresponding author: Anton P. Bonartsev, e-mail:
| | - V.V. Voinova
- Senior Researcher, Department of Biochemistry, Faculty of Biology; Lomonosov Moscow State University, 1–12 Leninskiye Gory, Moscow, 119234, Russia
| | - A.V. Volkov
- Senior Researcher; N.N. Priorov National Medical Research Center of Traumatology and Orthopedics, 10 Priorova St., Moscow, 127299, Russia; Associate Professor, Department of Pathological Anatomy, Medical Institute; Peoples’ Friendship University of Russia, 6 Miklukho-Maklaya St., Moscow, 117198, Russia
| | - A.A. Muraev
- Professor, Department of Maxillofacial Surgery and Surgical Dentistry; Peoples’ Friendship University of Russia, 6 Miklukho-Maklaya St., Moscow, 117198, Russia
| | - E.M. Boyko
- Teacher, Essentuki Branch; Stavropol State Medical University, 310 Mira St., Stavropol, 355017, Russia
| | - A.A. Venediktov
- Director; Cardioplant LLC, 1B Tsentralnaya St., Bldg. 2, Penza, 440004, Russia
| | - N.N. Didenko
- Assistant, Department of Pathological Physiology; Stavropol State Medical University, 310 Mira St., Stavropol, 355017, Russia
| | - A.A. Dolgalev
- Associate Professor, Professor, Department of General and Pediatric Dentistry; Stavropol State Medical University, 310 Mira St., Stavropol, 355017, Russia; Head of the Center for Innovation and Technology Transfer of the Research and Innovation Association; Stavropol State Medical University, 310 Mira St., Stavropol, 355017, Russia
| |
Collapse
|
7
|
Lima F, Teixeira M, Silva Júnior V, Costa A, Lima V, Souza H, Barros M, Graziano A, Naro F, Soares A. Avaliação da eficácia do sistema rigeneracon no tratamento de lesões de calvária em ratos. ARQ BRAS MED VET ZOO 2021. [DOI: 10.1590/1678-4162-11699] [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] Open
Abstract
RESUMO Defeitos ósseos constituem um problema de saúde global. O sistema Rigenera permite a extração de microenxertos ricos em células-tronco mesenquimais (CTMs). Objetivou-se avaliar o processo de regeneração óssea por enxertos obtidos pelo sistema Rigenera em defeitos críticos na calvária de ratos. Foram utilizados 18 ratos Wistar, machos, pesando 285±29g, distribuídos em três grupos (n=6), sendo cada animal controle de si mesmo, denominados G15-Controle e G15-Tratado (15 dias); G30-Controle e G30-Tratado (30 dias) e G60-Controle e G60-Tratado (60 dias). Foram realizadas duas lesões de 5mm de diâmetro em cada antímero da calvária. Nos grupos tratados, foram utilizados microenxertos autólogos de cartilagem xifoide, obtidos pelo sistema Rigenera. O defeito contralateral serviu como controle em todos os animais. Os animais foram eutanasiados aos 15, 30 e 60 dias após a cirurgia, e as amostras foram processadas para a histoquímica. Nos grupos controle, não foram observados sinais de regeneração óssea, enquanto nos grupos tratamento foram verificadas áreas de formação óssea e tecido mesenquimal ativado. O sistema Rigenera foi eficiente na obtenção de microenxertos autólogos, para terapia celular em defeito crítico de calvária de ratos. Com o aprimoramento do protocolo, o sistema Rigenera poderá ser amplamente utilizado no tratamento de lesões ósseas.
Collapse
Affiliation(s)
- F.C.S. Lima
- Universidade Federal Rural de Pernambuco, Brazil
| | | | | | - A.B.B. Costa
- Universidade Federal Rural de Pernambuco, Brazil
| | - V.C. Lima
- Universidade Federal Rural de Pernambuco, Brazil
| | - H.C.V. Souza
- Universidade Federal Rural de Pernambuco, Brazil
| | | | | | - F. Naro
- Sapienza University of Rome, Italy
| | - A.F. Soares
- Universidade Federal Rural de Pernambuco, Brazil
| |
Collapse
|
8
|
Okuchi Y, Reeves J, Ng SS, Doro DH, Junyent S, Liu KJ, El Haj AJ, Habib SJ. Wnt-modified materials mediate asymmetric stem cell division to direct human osteogenic tissue formation for bone repair. NATURE MATERIALS 2021; 20:108-118. [PMID: 32958876 DOI: 10.1038/s41563-020-0786-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 07/27/2020] [Indexed: 06/11/2023]
Abstract
The maintenance of human skeletal stem cells (hSSCs) and their progeny in bone defects is a major challenge. Here, we report on a transplantable bandage containing a three-dimensional Wnt-induced osteogenic tissue model (WIOTM). This bandage facilitates the long-term viability of hSSCs (8 weeks) and their progeny, and enables bone repair in an in vivo mouse model of critical-sized calvarial defects. The newly forming bone is structurally comparable to mature cortical bone and consists of human and murine cells. Furthermore, we show that the mechanism of WIOTM formation is governed by Wnt-mediated asymmetric cell division of hSSCs. Covalently immobilizing Wnts onto synthetic materials can polarize single dividing hSSCs, orient the spindle and simultaneously generate a Wnt-proximal hSSC and a differentiation-prone Wnt-distal cell. Our results provide insight into the regulation of human osteogenesis and represent a promising approach to deliver human osteogenic constructs that can survive in vivo and contribute to bone repair.
Collapse
Affiliation(s)
- Yoshihisa Okuchi
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
| | - Joshua Reeves
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
| | - Soon Seng Ng
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
| | - Daniel H Doro
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Sergi Junyent
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
| | - Karen J Liu
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Alicia J El Haj
- Healthcare Technology Institute, Institute of Translational Medicine, University of Birmingham, Birmingham, UK
| | - Shukry J Habib
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK.
| |
Collapse
|
9
|
Taguchi T, Lopez MJ. An overview of de novo bone generation in animal models. J Orthop Res 2021; 39:7-21. [PMID: 32910496 PMCID: PMC7820991 DOI: 10.1002/jor.24852] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 08/27/2020] [Accepted: 09/02/2020] [Indexed: 02/04/2023]
Abstract
Some of the earliest success in de novo tissue generation was in bone tissue, and advances, facilitated by the use of endogenous and exogenous progenitor cells, continue unabated. The concept of one health promotes shared discoveries among medical disciplines to overcome health challenges that afflict numerous species. Carefully selected animal models are vital to development and translation of targeted therapies that improve the health and well-being of humans and animals alike. While inherent differences among species limit direct translation of scientific knowledge between them, rapid progress in ex vivo and in vivo de novo tissue generation is propelling revolutionary innovation to reality among all musculoskeletal specialties. This review contains a comparison of bone deposition among species and descriptions of animal models of bone restoration designed to replicate a multitude of bone injuries and pathology, including impaired osteogenic capacity.
Collapse
Affiliation(s)
- Takashi Taguchi
- Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary MedicineLouisiana State UniversityBaton RougeLouisianaUSA
| | - Mandi J. Lopez
- Laboratory for Equine and Comparative Orthopedic Research, Department of Veterinary Clinical Sciences, School of Veterinary MedicineLouisiana State UniversityBaton RougeLouisianaUSA
| |
Collapse
|
10
|
Jensen MB, Slots C, Ditzel N, Kolstrup S, Kassem M, Thygesen T, Andersen MØ. Treating mouse skull defects with 3D-printed fatty acid and tricalcium phosphate implants. J Tissue Eng Regen Med 2020; 14:1858-1868. [PMID: 33098263 DOI: 10.1002/term.3146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 09/17/2020] [Accepted: 09/28/2020] [Indexed: 11/09/2022]
Abstract
Skull surgery, also known as craniectomy, is done to treat trauma or brain diseases and may require the use of an implant to reestablish skull integrity. This study investigates the performance of 3D printed bone implants in a mouse model of craniectomy with the aim of making biodegradable porous implants that can ultimately be fitted to a patient's anatomy. A nonpolymeric thermoplastic bioink composed of fatty acids and β-tricalcium phosphate was used to 3D print the skull implants. Some of these were sintered to yield pure β-tricalcium phosphate implants. The performance of nonsintered and sintered implants was then compared in two semi-quantitative murine calvarial defect models using computed tomography, histology, and luciferase activity. Both types of implants were biocompatible, but only sintered implants promoted defect healing, with osseointegration to adjacent bone and the formation of new bone and bone marrow tissue in the implant pores. Luciferase scanning and histology showed that mesenchymal stem cells seeded onto the implants engraft and proliferate on the implants after implantation and contribute to forming bone. The experiments indicate that fatty acid-based 3D printing enables the creation of biocompatible and bone-forming β-tricalcium phosphate implants.
Collapse
Affiliation(s)
- Martin Bonde Jensen
- Section for Biotechnology (SDU Biotechnology), Department of Green Technology, Faculty of Engineering, University of Southern Denmark, Odense, Denmark.,Particle3D ApS, Odense, Denmark
| | - Casper Slots
- Section for Biotechnology (SDU Biotechnology), Department of Green Technology, Faculty of Engineering, University of Southern Denmark, Odense, Denmark.,Particle3D ApS, Odense, Denmark
| | - Nicholas Ditzel
- Department of Endocrinology and Metabolism, Molecular Endocrinology Laboratory (KMEB), Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Stefanie Kolstrup
- The Biomedical Laboratory, University of Southern Denmark, Odense, Denmark
| | - Moustapha Kassem
- Department of Endocrinology and Metabolism, Molecular Endocrinology Laboratory (KMEB), Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Torben Thygesen
- Department of Oral and Maxillofacial Surgery, Odense University Hospital, Odense, Denmark
| | - Morten Østergaard Andersen
- Section for Biotechnology (SDU Biotechnology), Department of Green Technology, Faculty of Engineering, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
11
|
Aghali A, Arman HE. Photoencapsulated-mesenchymal stromal cells in biodegradable thiol-acrylate hydrogels enhance regeneration of craniofacial bone tissue defects. Regen Med 2020; 15:2115-2127. [PMID: 33211632 DOI: 10.2217/rme-2020-0061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 10/20/2020] [Indexed: 01/09/2023] Open
Abstract
Aim: This study investigated biodegradable thiol-acrylate hydrogels as stem cell carriers to facilitate cranial bone regeneration. Materials & methods: Two formulations of thiol-acrylate hydrogels (5 and 15 wt% Poly[ethylene glycol]-diacrylate [PEGDA] hydrogels) were used as stem cell carriers. Bone marrow mesenchymal stromal cells and dental pulp mesenchymal stromal cells were photoencapsulated and cultured in basal or osteogenic medium 3 days before the surgery. Using New Zealand White Rabbits, four defects (5 mm diameter and 2 mm thickness) were created and hydrogel scaffolds were implanted in each rabbit cranium for 6 weeks. Results & Conclusion: AlamarBlue assay showed increasing metabolic activity levels in 5 wt% PEGDA hydrogels than 15 wt% PEGDA hydrogels. Photoencapsulated-mesenchymal stromal cells in 15 wt% PEGDA hydrogels demonstrated significantly increasing alkaline phosphatase activity levels on day 7 compared with days 1 and 3. Histological diagnosis showed 5 wt% PEGDA hydrogels resulted in lower averaged residual gel areas than 15 wt% PEGDA hydrogel specimens and control groups 6 weeks postimplantation.
Collapse
Affiliation(s)
- Arbi Aghali
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55902, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47908, USA
| | - Huseyin E Arman
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
12
|
Ghaffarinovin Z, Soltaninia O, Mortazavi Y, Esmaeilzadeh A, Nadri S. Repair of rat cranial bone defect by using amniotic fluid-derived mesenchymal stem cells in polycaprolactone fibrous scaffolds and platelet-rich plasma. ACTA ACUST UNITED AC 2020; 11:209-217. [PMID: 34336609 PMCID: PMC8314035 DOI: 10.34172/bi.2021.28] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/20/2020] [Accepted: 05/10/2020] [Indexed: 12/28/2022]
Abstract
Introduction: Tissue regenerative medicine strategies, as a promising alternative has become of major interest to the reconstruction of critical size bone defects. This study evaluated the effects of the simultaneous application of polycaprolactone (PCL), amniotic fluid mesenchymal stem cells (AF-MSCs) and platelet-rich plasma (PRP) on the repair of rat cranial bone defects. Methods: The AF-MSCs were isolated at the end of the second week of pregnancy in rats. PRP obtained from rat blood and the random PCL fibrous scaffolds were prepared using the electrospinning method. Circular full thickness (5 mm) bone defects were developed on both sides of the parietal bones (animal number=24) and the scaffolds containing AF-MSCs and PRP were implanted in the right lesions. Thereafter, after eight weeks the histological and immunohistochemistry studies were performed to evaluate the bone formation and collagen type I expression. Results: The spindle-shaped mesenchymal stem cells were isolated and the electron microscope images indicated the preparation of a random PCL scaffold. Immunohistochemical findings showed that collagen type I was expressed by AF-MSCs cultured on the scaffold. The results of hematoxylin and eosin (H&E) staining indicated the formation of blood vessels in the presence of PRP. Additionally, immunofluorescence findings suggested that PRP had a positive effect on collagen type I expression. Conclusion: The simultaneous application of fibrous scaffold + AF-MSCs + PRP has positive effects on bone regeneration. This study showed that PRP can affect the formation of new blood vessels in the scaffold transplanted in the bone defect.
Collapse
Affiliation(s)
- Zeinab Ghaffarinovin
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Omid Soltaninia
- Department of Oral & Maxillofacial Surgery, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Yousef Mortazavi
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.,Cancer Gene therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Abdolreza Esmaeilzadeh
- Cancer Gene therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.,Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Samad Nadri
- Cancer Gene therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.,Department of Medical Nanotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.,Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.,Zanjan Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
13
|
Yu L, Rowe DW, Perera IP, Zhang J, Suib SL, Xin X, Wei M. Intrafibrillar Mineralized Collagen-Hydroxyapatite-Based Scaffolds for Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2020; 12:18235-18249. [PMID: 32212615 DOI: 10.1021/acsami.0c00275] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
As one of the major challenges in the field of tissue engineering, large skeletal defects have attracted wide attention from researchers. Collagen (Col) and hydroxyapatite (HA), the most abundant protein and the main component in natural bone, respectively, are usually used as a biomimetic composite material in tissue engineering due to their excellent biocompatibility and biodegradability. In this study, novel intrafibrillar mineralized Col-HA-based scaffolds, constructed in either cellular or lamellar microstructures, were established through a biomimetic method to enhance the new bone-regenerating capability of tissue engineering scaffolds. Moreover, iron (Fe) and manganese (Mn), two of the essential trace elements in the body, were successfully incorporated into the lamellar scaffold to further improve the osteoinductivity of these biomaterials. It was found that the lamellar scaffolds demonstrated better osteogenic abilities compared to both in-house and commercial Col-HA-based cellular scaffolds in vitro and in vivo. Meanwhile, Fe/Mn incorporation further amplified the osteogenic promotion of the lamellar scaffolds. More importantly, a synergistic effect was observed in the Fe and Mn dual-element-incorporated lamellar scaffolds for both in vitro osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and in vivo bone regeneration loaded with fresh bone marrow cells. This study provides a simple but practical strategy for the creation of functional scaffolds for bone regeneration.
Collapse
Affiliation(s)
- Le Yu
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, Ohio 45701, United States
| | - David W Rowe
- Center for Regenerative Medicine and Skeletal Development, School of Dental Medicine, University of Connecticut Health Center, Farmington, Connecticut 06032, United States
| | | | | | | | - Xiaonan Xin
- Center for Regenerative Medicine and Skeletal Development, School of Dental Medicine, University of Connecticut Health Center, Farmington, Connecticut 06032, United States
| | - Mei Wei
- Department of Mechanical Engineering, Ohio University, Athens, Ohio 45701, United States
| |
Collapse
|
14
|
Human Bone Marrow Mesenchymal Stromal Cells Promote Bone Regeneration in a Xenogeneic Rabbit Model: A Preclinical Study. Stem Cells Int 2018; 2018:7089484. [PMID: 30123292 PMCID: PMC6079361 DOI: 10.1155/2018/7089484] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 05/07/2018] [Accepted: 05/23/2018] [Indexed: 01/14/2023] Open
Abstract
Significant research efforts have been undertaken during the last decades to treat musculoskeletal disorders and improve patient's mobility and quality of life. The goal is the return of function as quickly and completely as possible. Cellular therapy has been increasingly employed in this setting. The design of this study was focused on cell-based alternatives. The present study aimed at investigating the bone regeneration capacity of xenogeneic human bone marrow-derived mesenchymal stromal cell (hMSC) implantation with tricalcium phosphate (TCP) granules in an immunocompetent rabbit model of critical-size bone defects at the femoral condyles. Two experimental groups, TCP and hMSC + TCP, were compared. Combination of TCP and hMSC did not affect cell viability or osteogenic differentiation. We also observed significantly higher bone regeneration in vivo in the hMSC + TCP group, which also displayed better TCP osteointegration. Also, evidence of hMSC contribution to a better TCP osteointegration was noticed. Finally, no inflammatory reaction was detected, besides the xenotransplantation of human cells into an immunocompetent recipient. In summary, hMSC combined with TCP granules is a potential combination for bone regeneration purposes that provides better preclinical results compared to TCP alone.
Collapse
|
15
|
Ndong JDLC, Stephenson Y, Davis ME, García AJ, Goudy S. Controlled JAGGED1 delivery induces human embryonic palate mesenchymal cells to form osteoblasts. J Biomed Mater Res A 2017; 106:552-560. [PMID: 28913955 DOI: 10.1002/jbm.a.36236] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/09/2017] [Accepted: 08/25/2017] [Indexed: 12/12/2022]
Abstract
Osteoblast commitment and differentiation are controlled by multiple growth factors including members of the Notch signaling pathway. JAGGED1 is a cell surface ligand of the Notch pathway that is necessary for murine bone formation. The delivery of JAGGED1 to induce bone formation is complicated by its need to be presented in a bound form to allow for proper Notch receptor signaling. In this study, we investigate whether the sustained release of JAGGED1 stimulates human mesenchymal cells to commit to osteoblast cell fate using polyethylene glycol malemeide (PEG-MAL) hydrogel delivery system. Our data demonstrated that PEG-MAL hydrogel constructs are stable in culture for at least three weeks and maintain human mesenchymal cell viability with little cytotoxicity in vitro. JAGGED1 loaded on PEG-MAL hydrogel (JAGGED1-PEG-MAL) showed continuous release from the gel for up to three weeks, with induction of Notch signaling using a CHO cell line with a Notch1 reporter construct, and qPCR gene expression analysis in vitro. Importantly, JAGGED1-PEG-MAL hydrogel induced mesenchymal cells towards osteogenic differentiation based on increased Alkaline phosphatase activity and osteoblast genes expression including RUNX2, ALP, COL1, and BSP. These results thus indicated that JAGGED1 delivery in vitro using PEG-MAL hydrogel induced osteoblast commitment, suggesting that this may be a viable in vivo approach to bone regeneration. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 552-560, 2018.
Collapse
Affiliation(s)
| | | | - Michael E Davis
- Wallace H. Coulter Department of Biomedical Engineering at Emory University and Georgia Institute of Technology, Atlanta, Georgia
| | - Andrés J García
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia.,Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
| | - Steven Goudy
- Emory University, Department of Otolaryngology, Atlanta, Georgia.,Emory University, Department of Pediatrics, Atlanta, Georgia
| |
Collapse
|
16
|
Murphy MP, Quarto N, Longaker MT, Wan DC. * Calvarial Defects: Cell-Based Reconstructive Strategies in the Murine Model. Tissue Eng Part C Methods 2017; 23:971-981. [PMID: 28825366 DOI: 10.1089/ten.tec.2017.0230] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Calvarial defects pose a continued clinical dilemma for reconstruction. Advancements within the fields of stem cell biology and tissue engineering have enabled researchers to develop reconstructive strategies using animal models. We review the utility of various animal models and focus on the mouse, which has aided investigators in understanding cranial development and calvarial bone healing. The murine model has also been used to study regenerative approaches to critical-sized calvarial defects, and we discuss the application of stem cells such as bone marrow-derived mesenchymal stromal cells, adipose-derived stromal cells, muscle-derived stem cells, and pluripotent stem cells to address deficient bone in this animal. Finally, we highlight strategies to manipulate stem cells using various growth factors and inhibitors and ultimately how these factors may prove crucial in future advancements within calvarial reconstruction using native skeletal stem cells.
Collapse
Affiliation(s)
- Matthew P Murphy
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University , Stanford, California.,2 Lorry I. Lokey Stem Cell Research Building, Stanford Stem Cell Biology and Regenerative Medicine Institute, Stanford University , Stanford, California
| | - Natalina Quarto
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University , Stanford, California
| | - Michael T Longaker
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University , Stanford, California.,2 Lorry I. Lokey Stem Cell Research Building, Stanford Stem Cell Biology and Regenerative Medicine Institute, Stanford University , Stanford, California
| | - Derrick C Wan
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University , Stanford, California
| |
Collapse
|
17
|
Li J, Huang Z, Chen L, Tang X, Fang Y, Liu L. Restoration of bone defects using modified heterogeneous deproteinized bone seeded with bone marrow mesenchymal stem cells. Am J Transl Res 2017; 9:3200-3211. [PMID: 28804540 PMCID: PMC5553872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 06/20/2017] [Indexed: 06/07/2023]
Abstract
The aim of the present study was to investigate the effect of modified heterogeneous deproteinized bone combined with bone marrow mesenchymal stem cells (BMSCs) in the restoration of a validated bone defect model. BMSCs were identified by flow cytometry and multilineage differentiation assay. The structural features of the modified heterogeneous deproteinized bone scaffold and biocompatibility between BMSCs and the scaffold were confirmed by scanning electron microscope (SEM) detection. The cytotoxicity of the modified heterogeneous deproteinized bone scaffolds were detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenytetrazolium bromide (MTT) assay. SEM detection proved that modified heterogeneous deproteinized bone scaffold had no negative impact on the proliferation of BMSCs. MTT assay results demonstrated that the scaffold had no apparent cytotoxicity. Biomechanical detection showed that the stiffness and ultimate loading of tibias in the scaffold + BMSCs group were significantly higher than those of the scaffold alone group (P < 0.05) and the control group (P < 0.01). Histological analyses confirmed that the greatest quantity of new bone was generated in the scaffold + BMSCs group, when compared with all other groups, at 8 weeks' post-operation. The bone mineral density (BMD) in the scaffold + BMSC group was significantly higher than that of the scaffold alone group (P < 0.05) and the control group (P < 0.01). Fluorometric analyses confirmed the presence of BMSCs at high concentration within the bone defect areas in the scaffold + BMSCs group at 4 weeks after transplantation. These findings suggest that the modified heterogeneous deproteinized bone scaffold seeded with BMSCs can effectively enhance the restoration of bone defects.
Collapse
Affiliation(s)
- Jun Li
- Department of Orthopaedics, West China Hospital, Sichuan University37# Wainan Guoxue Road, Chengdu 610041, People's Republic of China
| | - Zeyu Huang
- Department of Orthopaedics, West China Hospital, Sichuan University37# Wainan Guoxue Road, Chengdu 610041, People's Republic of China
| | - Liyan Chen
- Department of Orthopaedics, West China Hospital, Sichuan University37# Wainan Guoxue Road, Chengdu 610041, People's Republic of China
| | - Xin Tang
- Department of Orthopaedics, West China Hospital, Sichuan University37# Wainan Guoxue Road, Chengdu 610041, People's Republic of China
| | - Yue Fang
- Department of Orthopaedics, West China Hospital, Sichuan University37# Wainan Guoxue Road, Chengdu 610041, People's Republic of China
| | - Lei Liu
- Department of Orthopaedics, West China Hospital, Sichuan University37# Wainan Guoxue Road, Chengdu 610041, People's Republic of China
| |
Collapse
|
18
|
Abstract
Unlike many other postnatal tissues, bone can regenerate and repair itself; nevertheless, this capacity can be overcome. Traditionally, surgical reconstructive strategies have implemented autologous, allogeneic, and prosthetic materials. Autologous bone--the best option--is limited in supply and also mandates an additional surgical procedure. In regenerative tissue engineering, there are myriad issues to consider in the creation of a functional, implantable replacement tissue. Importantly, there must exist an easily accessible, abundant cell source with the capacity to express the phenotype of the desired tissue, and a biocompatible scaffold to deliver the cells to the damaged region. A literature review was performed using PubMed; peer-reviewed publications were screened for relevance in order to identify key advances in stem and progenitor cell contribution to the field of bone tissue engineering. In this review, we briefly introduce various adult stem cells implemented in bone tissue engineering such as mesenchymal stem cells (including bone marrow- and adipose-derived stem cells), endothelial progenitor cells, and induced pluripotent stem cells. We then discuss numerous advances associated with their application and subsequently focus on technological advances in the field, before addressing key regenerative strategies currently used in clinical practice. Stem and progenitor cell implementation in bone tissue engineering strategies have the ability to make a major impact on regenerative medicine and reduce patient morbidity. As the field of regenerative medicine endeavors to harness the body's own cells for treatment, scientific innovation has led to great advances in stem cell-based therapies in the past decade.
Collapse
|
19
|
Sugamori Y, Mise-Omata S, Maeda C, Aoki S, Tabata Y, Murali R, Yasuda H, Udagawa N, Suzuki H, Honma M, Aoki K. Peptide drugs accelerate BMP-2-induced calvarial bone regeneration and stimulate osteoblast differentiation through mTORC1 signaling. Bioessays 2016; 38:717-25. [PMID: 27345003 PMCID: PMC5094554 DOI: 10.1002/bies.201600104] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Both W9 and OP3‐4 were known to bind the receptor activator of NF‐κB ligand (RANKL), inhibiting osteoclastogenesis. Recently, both peptides were shown to stimulate osteoblast differentiation; however, the mechanism underlying the activity of these peptides remains to be clarified. A primary osteoblast culture showed that rapamycin, an mTORC1 inhibitor, which was recently demonstrated to be an important serine/threonine kinase for bone formation, inhibited the peptide‐induced alkaline phosphatase activity. Furthermore, both peptides promoted the phosphorylation of Akt and S6K1, an upstream molecule of mTORC1 and the effector molecule of mTORC1, respectively. In the in vivo calvarial defect model, W9 and OP3‐4 accelerated BMP‐2‐induced bone formation to a similar extent, which was confirmed by histomorphometric analyses using fluorescence images of undecalcified sections. Our data suggest that these RANKL‐binding peptides could stimulate the mTORC1 activity, which might play a role in the acceleration of BMP‐2‐induced bone regeneration by the RANKL‐binding peptides.
Collapse
Affiliation(s)
- Yasutaka Sugamori
- Department of Bio-Matrix (Pharmacology), Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Setsuko Mise-Omata
- Department of Bio-Matrix (Pharmacology), Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Chizuko Maeda
- Department of Bio-Matrix (Pharmacology), Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shigeki Aoki
- Faculty of Medicine, Department of Pharmacy, The University of Tokyo Hospital, The University of Tokyo, Tokyo, Japan
| | - Yasuhiko Tabata
- Department of Biomaterials, Field of Tissue Engineering, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Ramachandran Murali
- Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hisataka Yasuda
- Nagahama Institute for Biochemical Science, Oriental Yeast Co. Ltd., Shiga, Japan
| | - Nobuyuki Udagawa
- Department of Biochemistry, Matsumoto Dental University, Nagano, Japan
| | - Hiroshi Suzuki
- Faculty of Medicine, Department of Pharmacy, The University of Tokyo Hospital, The University of Tokyo, Tokyo, Japan
| | - Masashi Honma
- Faculty of Medicine, Department of Pharmacy, The University of Tokyo Hospital, The University of Tokyo, Tokyo, Japan.,Faculty of Medicine, Department of Pharmacology and Pharmacokinetics, The University of Tokyo Hospital, The University of Tokyo, Tokyo, Japan
| | - Kazuhiro Aoki
- Department of Bio-Matrix (Pharmacology), Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
20
|
Lee ES, Park J, Wang J, Lee H, Hwang NS. Osteogenic commitment of human induced pluripotent stem cell-derived mesenchymal progenitor-like cells on biomimetic scaffolds. J IND ENG CHEM 2016. [DOI: 10.1016/j.jiec.2016.03.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
21
|
Yue X, Niu M, Zhang T, Wang C, Wang Z, Wu W, Zhang Q, Lai C, Zhou L. In vivo evaluation of a simvastatin-loaded nanostructured lipid carrier for bone tissue regeneration. NANOTECHNOLOGY 2016; 27:115708. [PMID: 26881419 DOI: 10.1088/0957-4484/27/11/115708] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Alveolar bone loss has long been a challenge in clinical dental implant therapy. Simvastatin (SV) has been demonstrated to exert excellent anabolic effects on bone. However, the successful use of SV to increase bone formation in vivo largely depends on the local concentration of SV at the site of action, and there have been continuing efforts to develop an appropriate delivery system. Specifically, nanostructured lipid carrier (NLC) systems have become a popular type of encapsulation carrier system. Therefore, SV-loaded NLCs (SNs) (179.4 nm in diameter) were fabricated in this study, and the osteogenic effect of the SNs was evaluated in a critical-sized rabbit calvarial defect. Our results revealed that the SNs significantly enhanced bone formation in vivo, as evaluated by hematoxylin and eosin (HE) staining, immunohistochemistry, and a fluorescence analysis. Thus, this novel nanostructured carrier system could be a potential encapsulation carrier system for SV in bone regeneration applications.
Collapse
Affiliation(s)
- Xinxin Yue
- Center of Oral Implantology, Guangdong Provincial Stomatological Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Overcoming translational challenges - The delivery of mechanical stimuli in vivo. Int J Biochem Cell Biol 2015; 69:162-72. [PMID: 26482595 DOI: 10.1016/j.biocel.2015.10.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 10/11/2015] [Accepted: 10/12/2015] [Indexed: 01/22/2023]
Abstract
Despite major medical advances, non-union bone fractures and skeletal defects continue to place significant burden on the patient, the clinicians and the healthcare system as a whole. Current bone substitute approaches are still limited in effectiveness and to date no adequate bone substitute material has been developed for routine clinical application. Tissue engineering presents a novel approach to tackling this clinical burden and developing an acceptable solution for the treatment of skeletal defects. Over the past three decades the field has evolved to appreciate the key biological, material and physical parameters influencing the development of a cell-based tissue engineered therapy and to create associated technologies to exploit such parameters. In recent years a number of therapies have started progressing along the pre-clinical pipeline to build a case for regulatory approval and ultimately clinical adoption. However, little emphasis has been given to the translational challenges faced when moving from "bench-to-bedside". One particular challenge lies in the delivery of functional mechanical stimuli to implanted cell populations to activate and promote osteogenic activities. This review introduces novel bio-magnetic approaches to overcoming this challenge.
Collapse
|
23
|
Interactions between MSCs and immune cells: implications for bone healing. J Immunol Res 2015; 2015:752510. [PMID: 26000315 PMCID: PMC4427002 DOI: 10.1155/2015/752510] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 01/18/2015] [Accepted: 01/19/2015] [Indexed: 02/07/2023] Open
Abstract
It is estimated that, of the 7.9 million fractures sustained in the United States each year, 5% to 20% result in delayed or impaired healing requiring therapeutic intervention. Following fracture injury, there is an initial inflammatory response that plays a crucial role in bone healing; however, prolonged inflammation is inhibitory for fracture repair. The precise spatial and temporal impact of immune cells and their cytokines on fracture healing remains obscure. Some cytokines are reported to be proosteogenic while others inhibit bone healing. Cell-based therapy utilizing mesenchymal stromal cells (MSCs) is an attractive option for augmenting the fracture repair process. Osteoprogenitor MSCs not only differentiate into bone, but they also exert modulatory effects on immune cells via a variety of mechanisms. In this paper, we review the current literature on both in vitro and in vivo studies on the role of the immune system in fracture repair, the use of MSCs in the enhancement of fracture healing, and interactions between MSCs and immune cells. Insight into this paradigm can provide valuable clues in identifying cellular and noncellular targets that can potentially be modulated to enhance both natural bone healing and bone repair augmented by the exogenous addition of MSCs.
Collapse
|
24
|
Lei Q, Chen J, Huang W, Wu D, Lin H, Lai Y. Proteomic analysis of the effect of extracellular calcium ions on human mesenchymal stem cells: Implications for bone tissue engineering. Chem Biol Interact 2015; 233:139-46. [PMID: 25824407 DOI: 10.1016/j.cbi.2015.03.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 01/29/2015] [Accepted: 03/19/2015] [Indexed: 12/16/2022]
Abstract
Human mesenchymal stem cells-bone marrow (BM-hMSCs) are considered as the most suitable seed cells for bone tissue engineering. Calcium ions (Ca(2+)) forms an important component of a number of commercial bone substitutes and support materials. For efficient bone tissue engineering, it is crucial to explore the effect of extracellular Ca(2+) on the growth and differentiation of BM-hMSCs, and to understand their molecular mechanisms. Therefore, in the present study, BM-hMSCs were cultivated in serum free growth medium or serum free growth medium with additional 4 or 6mM Ca(2+) for 3weeks, following which, the proliferation and osteoblastic differentiation of these cells were evaluated. Differentially expressed proteins were established using iTRAQ labeling coupled with nano-LC-MS/MS. Our data revealed that Ca(2+) significantly promoted the proliferation of BM-hMSCs in the early stage. Furthermore, Ca(2+) showed osteoinduction properties. MAPKs signaling pathway might participate in the osteogenic differentiation of BM-hMSCs caused by Ca(2+). Certain newly found proteins could be potentially important for the osteogenic differentiation of BM-hMSCs and may be associated with osteogenesis.
Collapse
Affiliation(s)
- Qun Lei
- Department of Oral Implantology, Affiliated Stomatological Hospital of Fujian Medical University, Fuzhou, Fujian 350002, China
| | - Jiang Chen
- Department of Oral Implantology, Affiliated Stomatological Hospital of Fujian Medical University, Fuzhou, Fujian 350002, China
| | - Wenxiu Huang
- Department of Oral Implantology, Affiliated Stomatological Hospital of Fujian Medical University, Fuzhou, Fujian 350002, China
| | - Dong Wu
- Department of Oral Implantology, Affiliated Stomatological Hospital of Fujian Medical University, Fuzhou, Fujian 350002, China.
| | - Hengzhang Lin
- Department of Stomatology, Affiliated Provincial Governmental Hospital of Fujian Health College, Fuzhou, Fujian 350002, China
| | - Yingzhen Lai
- Department of Stomatology, Xiamen Medical College, Xiamen, Fujian 361008, China
| |
Collapse
|
25
|
Frohbergh ME, Katsman A, Mondrinos MJ, Stabler CT, Hankenson KD, Oristaglio JT, Lelkes PI. Osseointegrative properties of electrospun hydroxyapatite-containing nanofibrous chitosan scaffolds. Tissue Eng Part A 2014; 21:970-81. [PMID: 25336062 DOI: 10.1089/ten.tea.2013.0789] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Our long-term goal is to develop smart biomaterials that can facilitate regeneration of critical-size craniofacial lesions. In this study, we tested the hypothesis that biomimetic scaffolds electrospun from chitosan (CTS) will promote tissue repair and regeneration in a critical size calvarial defect. To test this hypothesis, we first compared in vitro ability of electrospun CTS scaffolds crosslinked with genipin (CTS-GP) to those of mineralized CTS-GP scaffolds containing hydroxyapatite (CTS-HA-GP), by assessing proliferation/metabolic activity and alkaline phosphatase (ALP) levels of murine mesenchymal stem cells (mMSCs). The cells' metabolic activity exhibited a biphasic behavior, indicative of initial proliferation followed by subsequent differentiation for all scaffolds. ALP activity of mMSCs, a surrogate measure of osteogenic differentiation, increased over time in culture. After 3 weeks in maintenance medium, ALP activity of mMSCs seeded onto CTS-HA-GP scaffolds was approximately two times higher than that of cells cultured on CTS-GP scaffolds. The mineralized CTS-HA-GP scaffolds were also osseointegrative in vivo, as inferred from the enhanced bone regeneration in a murine model of critical size calvarial defects. Tissue regeneration was evaluated over a 3 month period by microCT and histology (Hematoxylin and Eosin and Masson's Trichrome). Treatment of the lesions with CTS-HA-GP scaffolds induced a 38% increase in the area of de novo generated mineralized tissue area after 3 months, whereas CTS-GP scaffolds only led to a 10% increase. Preseeding with mMSCs significantly enhanced the regenerative capacity of CTS-GP scaffolds (by ∼3-fold), to 35% increase in mineralized tissue area after 3 months. CTS-HA-GP scaffolds preseeded with mMSCs yielded 45% new mineralized tissue formation in the defects. We conclude that the presence of HA in the CTS-GP scaffolds significantly enhances their osseointegrative capacity and that mineralized chitosan-based scaffolds crosslinked with genipin may represent a unique biomaterial with possible clinical relevance for the repair of critical calvarial bone defects.
Collapse
Affiliation(s)
- Michael E Frohbergh
- 1 School of Biomedical Engineering, Science and Health Systems, Drexel University , Philadelphia, Pennsylvania
| | | | | | | | | | | | | |
Collapse
|
26
|
The effect of simvastatin on chemotactic capability of SDF-1α and the promotion of bone regeneration. Biomaterials 2014; 35:4489-98. [DOI: 10.1016/j.biomaterials.2014.02.025] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 02/13/2014] [Indexed: 12/29/2022]
|