1
|
Arunjaroensuk S, Thunyakitpisal P, Nampuksa K, Monmaturapoj N, Mattheos N, Pimkhaokham A. Stability of guided bone regeneration with two ratios of biphasic calcium phosphate at implant sites in the esthetic zone: A randomized controlled clinical trial. Clin Oral Implants Res 2023; 34:850-862. [PMID: 37314107 DOI: 10.1111/clr.14113] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 05/23/2023] [Accepted: 05/30/2023] [Indexed: 06/15/2023]
Abstract
AIMS The aim of this randomized, double-blind, clinical trial was to compare the stability of the horizontal dimensions (facial bone thickness) of augmented bone using biphasic calcium phosphate (BCP) with hydroxyapatite/β-tricalcium phosphate ratio of either 60/40 or 70/30. MATERIALS AND METHODS Sixty dental implants placed with contour augmentation in the esthetic zone were randomized to 60/40 BCP (n = 30) or 70/30 BCP (n = 30). Cone-beam computed tomographic was used to assess facial bone thickness post-implantation and 6 months later at implant platform and 2, 4, and 6 mm apical to it. RESULTS The percentage of horizontal dimension reduction was 23.64%, 12.83%, 9.62%, and 8.21% in 70/30 BCP group, while 44.26%, 31.91%, 25.88%, and 21.49% in 60/40 BCP group at the level of the implant platform and 2, 4, and 6 mm apical, respectively. Statistically significant difference was found at 6 months at all levels of measurement (p-value < .05). CONCLUSIONS BCP bone grafts with HA/β-TCP ratio of 60/40 and 70/30 showed comparable outcomes for contour augmentation simultaneously with implant placement. Interestingly, the 70/30 ratio was significantly superior in maintaining facial thickness and showed more stable horizontal dimensions of the augmented site.
Collapse
Affiliation(s)
- Sirida Arunjaroensuk
- Dental Biomaterials Science Program, Graduate School, Chulalongkorn University, Bangkok, Thailand
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Pasutha Thunyakitpisal
- Research Unit of Herbal Medicine, Biomaterials and Materials for Dental Treatment, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Katanchalee Nampuksa
- Assistive Technology and Medical Devices Research Center (A-MED), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Naruporn Monmaturapoj
- Assistive Technology and Medical Devices Research Center (A-MED), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Nikos Mattheos
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Department of Dental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Atiphan Pimkhaokham
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
2
|
Creste CFZ, Orsi PR, Landim-Alvarenga FC, Justulin LA, Golim MDA, Barraviera B, Ferreira RS. Highly effective fibrin biopolymer scaffold for stem cells upgrading bone regeneration. MATERIALS 2020; 13:ma13122747. [PMID: 32560388 PMCID: PMC7344939 DOI: 10.3390/ma13122747] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 05/29/2020] [Accepted: 06/05/2020] [Indexed: 12/19/2022]
Abstract
Fibrin scaffold fits as a provisional platform promoting cell migration and proliferation, angiogenesis, connective tissue formation and growth factors stimulation. We evaluated a unique heterologous fibrin biopolymer as scaffold to mesenchymal stem cells (MSCs) to treat a critical-size bone defect. Femurs of 27 rats were treated with fibrin biopolymer (FBP); FBP + MSCs; and FBP + MSC differentiated in bone lineage (MSC-D). Bone repair was evaluated 03, 21 and 42 days later by radiographic, histological and scanning electron microscopy (SEM) imaging. The FBP + MSC-D association was the most effective treatment, since newly formed Bone was more abundant and early matured in just 21 days. We concluded that FBP is an excellent scaffold for MSCs and also use of differentiated cells should be encouraged in regenerative therapy researches. The FBP ability to maintain viable MSCs at Bone defect site has modified inflammatory environment and accelerating their regeneration.
Collapse
Affiliation(s)
- Camila Fernanda Zorzella Creste
- Center for the Study of Venoms and Venomous Animals (CEVAP), UNESP—São Paulo State University, Botucatu 18610-307, Brazil; (C.F.Z.C.); (P.R.O.); (B.B.)
- Botucatu Medical School, UNESP—São Paulo State University, Botucatu 18618-687, Brazil;
| | - Patrícia Rodrigues Orsi
- Center for the Study of Venoms and Venomous Animals (CEVAP), UNESP—São Paulo State University, Botucatu 18610-307, Brazil; (C.F.Z.C.); (P.R.O.); (B.B.)
| | - Fernanda Cruz Landim-Alvarenga
- College of Veterinary Medicine and Animal Husbandry (FMVZ), UNESP—São Paulo State University, Botucatu 18618-681, Brazil;
| | - Luis Antônio Justulin
- Botucatu Biosciences Institute, UNESP—São Paulo State University, Botucatu 18618-689, Brazil;
| | | | - Benedito Barraviera
- Center for the Study of Venoms and Venomous Animals (CEVAP), UNESP—São Paulo State University, Botucatu 18610-307, Brazil; (C.F.Z.C.); (P.R.O.); (B.B.)
- Botucatu Medical School, UNESP—São Paulo State University, Botucatu 18618-687, Brazil;
| | - Rui Seabra Ferreira
- Center for the Study of Venoms and Venomous Animals (CEVAP), UNESP—São Paulo State University, Botucatu 18610-307, Brazil; (C.F.Z.C.); (P.R.O.); (B.B.)
- Botucatu Medical School, UNESP—São Paulo State University, Botucatu 18618-687, Brazil;
- Correspondence: ; Tel.: +55-(014)-3880-7241
| |
Collapse
|
3
|
Dupard SJ, Grigoryan A, Farhat S, Coutu DL, Bourgine PE. Development of Humanized Ossicles: Bridging the Hematopoietic Gap. Trends Mol Med 2020; 26:552-569. [PMID: 32470383 DOI: 10.1016/j.molmed.2020.01.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/16/2020] [Accepted: 01/31/2020] [Indexed: 02/06/2023]
Abstract
Ectopic 'humanized ossicles' (hOss) are miniaturized, engineered human bone organs in mice displaying a similar structure and function to native mouse bones. However, they are composed of human mesenchymal derived cells forming a humanized bone marrow niche. This in vivo reconstitution of human skeletal and hematopoietic compartments provides an opportunity to investigate the cellular and molecular processes involved in their establishment and functions in a human setting. However, current hOs strategies vary in their engineering methods and their downstream applications, undermining comprehensive exploitation of their potential. This review describes the specificities of the hOs models and highlights their potential and limits. Ultimately, we propose directions for the development of hOss as a technological platform for human hematopoietic studies.
Collapse
Affiliation(s)
- Steven J Dupard
- Laboratory for Cell, Tissue, and Organ engineering, Department of Clinical Sciences, Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Stem Cell Center, Lund University, Lund, Sweden
| | - Ani Grigoryan
- Laboratory for Cell, Tissue, and Organ engineering, Department of Clinical Sciences, Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Stem Cell Center, Lund University, Lund, Sweden
| | - Stephanie Farhat
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada; Division of Orthopedic Surgery, The Ottawa Hospital, Ottawa, ON, Canada
| | - Daniel L Coutu
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada; Division of Orthopedic Surgery, The Ottawa Hospital, Ottawa, ON, Canada
| | - Paul E Bourgine
- Laboratory for Cell, Tissue, and Organ engineering, Department of Clinical Sciences, Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Stem Cell Center, Lund University, Lund, Sweden.
| |
Collapse
|
4
|
Effects of Bone Marrow Stromal Cell Transplantation on Repair of Bone Defect in Rats. Trauma Mon 2018. [DOI: 10.5812/traumamon.13701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
5
|
Abarrategi A, Mian SA, Passaro D, Rouault-Pierre K, Grey W, Bonnet D. Modeling the human bone marrow niche in mice: From host bone marrow engraftment to bioengineering approaches. J Exp Med 2018; 215:729-743. [PMID: 29453226 PMCID: PMC5839768 DOI: 10.1084/jem.20172139] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 01/19/2018] [Accepted: 01/30/2018] [Indexed: 12/11/2022] Open
Abstract
Xenotransplantation of patient-derived samples in mouse models has been instrumental in depicting the role of hematopoietic stem and progenitor cells in the establishment as well as progression of hematological malignancies. The foundations for this field of research have been based on the development of immunodeficient mouse models, which provide normal and malignant human hematopoietic cells with a supportive microenvironment. Immunosuppressed and genetically modified mice expressing human growth factors were key milestones in patient-derived xenograft (PDX) models, highlighting the importance of developing humanized microenvironments. The latest major improvement has been the use of human bone marrow (BM) niche-forming cells to generate human-mouse chimeric BM tissues in PDXs, which can shed light on the interactions between human stroma and hematopoietic cells. Here, we summarize the methods used for human hematopoietic cell xenotransplantation and their milestones and review the latest approaches in generating humanized BM tissues in mice to study human normal and malignant hematopoiesis.
Collapse
Affiliation(s)
- Ander Abarrategi
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, England, UK
| | - Syed A Mian
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, England, UK
- Department of Haematological Medicine, King's College London School of Medicine, London, England, UK
| | - Diana Passaro
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, England, UK
| | - Kevin Rouault-Pierre
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, England, UK
- Department of Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, England, UK
| | - William Grey
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, England, UK
| | - Dominique Bonnet
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, England, UK
| |
Collapse
|
6
|
Jang SJ, Kim SE, Han TS, Son JS, Kang SS, Choi SH. Bone Regeneration of Hydroxyapatite with Granular Form or Porous Scaffold in Canine Alveolar Sockets. ACTA ACUST UNITED AC 2018; 31:335-341. [PMID: 28438860 DOI: 10.21873/invivo.11064] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/06/2017] [Accepted: 03/10/2017] [Indexed: 01/25/2023]
Abstract
This study was undertaken to assess bone regeneration using hydroxyapatite (HA). The primary focus was comparison of bone regeneration between granular HA (gHA) forms and porous HA (pHA) scaffold. The extracted canine alveolar sockets were divided with three groups: control, gHA and pHA. Osteogenic effect in the gHA and pHA groups showed bone-specific surface and bone mineral density to be significantly higher than that of the control group (p<0.01). Bone volume fraction, bone mineral density, and amount of connective tissue related to disturbing osseointegration of the gHA group was higher than in the pHA group. Quantity of new bone formation of the pHA group was higher than that of the gHA group. This study demonstrated that gHA and pHA are potentially good bone substitutes for alveolar socket healing. For new bone formation during 8 weeks' post-implantation, HA with porous scaffold was superior to the granular form of HA.
Collapse
Affiliation(s)
- Seok Jin Jang
- Department of Veterinary Surgery, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Se Eun Kim
- College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Tae Sung Han
- Department of Veterinary Surgery, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Jun Sik Son
- High-Tech Fiber R&D Headquarters, Korea Textile Development Institute, Daegu, Republic of Korea
| | - Seong Soo Kang
- College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Seok Hwa Choi
- Department of Veterinary Surgery, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|
7
|
Spalthoff S, Zimmerer R, Dittmann J, Kokemüller H, Tiede M, Flohr L, Korn P, Gellrich NC, Jehn P. Heterotopic bone formation in the musculus latissimus dorsi of sheep using β-tricalcium phosphate scaffolds: evaluation of different seeding techniques. Regen Biomater 2017; 5:77-84. [PMID: 29644089 PMCID: PMC5888254 DOI: 10.1093/rb/rbx029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 09/27/2017] [Accepted: 09/28/2017] [Indexed: 12/21/2022] Open
Abstract
Osseous reconstruction of large bone defects remains a challenge in oral and maxillofacial surgery. In addition to autogenous bone grafts, which despite potential donor-site mobility still represent the gold standard in reconstructive surgery, many studies have investigated less invasive alternatives such as in vitro cultivation techniques. This study compared different types of seeding techniques on pure β-tricalcium phosphate scaffolds in terms of bone formation and ceramic resorption in vivo. Cylindrical scaffolds loaded with autologous cancellous bone, venous blood, bone marrow aspirate concentrate or extracorporeal in vitro cultivated bone marrow stromal cells were cultured in sheep on a perforator vessel of the musculus latissimus dorsi over a 6-month period. Histological and histomorphometric analyses revealed that scaffolds loaded with cancellous bone were superior at promoting heterotopic bone formation and ceramic degradation, with autogenous bone and bone marrow aspirate concentrate inducing in vivo formation of vital bone tissue. These results confirm that autologous bone constitutes the preferred source of osteoinductive and osteogenic material that can reliably induce heterotopic bone formation in vivo.
Collapse
Affiliation(s)
- Simon Spalthoff
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany and
- Correspondence address. Department of Oral and Maxillofacial Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany. Tel: +49-511-532-4879; Fax: +49-511-532-18598; E-mail:
| | - Rüdiger Zimmerer
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany and
| | - Jan Dittmann
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany and
| | - Horst Kokemüller
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany and
| | - Marco Tiede
- Department of Conservative Dentistry, Periodontology and Preventive Dentistry, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Laura Flohr
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany and
| | - Philippe Korn
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany and
| | - Nils-Claudius Gellrich
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany and
| | - Philipp Jehn
- Department of Oral and Maxillofacial Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany and
| |
Collapse
|
8
|
Pérez-Campo FM, May T, Zauers J, Sañudo C, Delgado-Calle J, Arozamena J, Berciano MT, Lafarga M, Riancho JA. Generation and characterization of two immortalized human osteoblastic cell lines useful for epigenetic studies. J Bone Miner Metab 2017; 35:150-160. [PMID: 27038990 DOI: 10.1007/s00774-016-0753-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 02/22/2016] [Indexed: 10/22/2022]
Abstract
Different model systems using osteoblastic cell lines have been developed to help understand the process of bone formation. Here, we report the establishment of two human osteoblastic cell lines obtained from primary cultures upon transduction of immortalizing genes. The resulting cell lines had no major differences to their parental lines in their gene expression profiles. Similar to primary osteoblastic cells, osteocalcin transcription increased following 1,25-dihydroxyvitamin D3 treatment and the immortalized cells formed a mineralized matrix, as detected by Alizarin Red staining. Moreover, these human cell lines responded by upregulating ALPL gene expression after treatment with the demethylating agent 5-aza-2'-deoxycytidine (AzadC), as shown before for primary osteoblasts. We further demonstrate that these cell lines can differentiate in vivo, using a hydroxyapatite/tricalcium phosphate composite as a scaffold, to produce bone matrix. More importantly, we show that these cells respond to demethylating treatment, as shown by the increase in SOST mRNA levels, the gene encoding sclerostin, upon treatment of the recipient mice with AzadC. This also confirms, in vivo, the role of DNA methylation in the regulation of SOST expression previously shown in vitro. Altogether our results show that these immortalized cell lines constitute a particularly useful model system to obtain further insight into bone homeostasis, and particularly into the epigenetic mechanisms regulating sclerostin production.
Collapse
Affiliation(s)
- Flor M Pérez-Campo
- Department of Internal Medicine, Hospital U. Marqués de Valdecilla-IDIVAL Universidad de Cantabria, 39008, Santander, Cantabria, Spain
- Department of Molecular Biology, University of Cantabria, IDIVAL, Santander, Spain
| | | | | | - Carolina Sañudo
- Department of Internal Medicine, Hospital U. Marqués de Valdecilla-IDIVAL Universidad de Cantabria, 39008, Santander, Cantabria, Spain
| | - Jesús Delgado-Calle
- Department of Internal Medicine, Hospital U. Marqués de Valdecilla-IDIVAL Universidad de Cantabria, 39008, Santander, Cantabria, Spain
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Roudebush Veterans Administration Medical Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jana Arozamena
- Department of Internal Medicine, Hospital U. Marqués de Valdecilla-IDIVAL Universidad de Cantabria, 39008, Santander, Cantabria, Spain
| | - María T Berciano
- Department of Anatomy and Cell Biology, University of Cantabria, IDIVAL, Santander, Spain
| | - Miguel Lafarga
- Department of Anatomy and Cell Biology, University of Cantabria, IDIVAL, Santander, Spain
| | - José A Riancho
- Department of Internal Medicine, Hospital U. Marqués de Valdecilla-IDIVAL Universidad de Cantabria, 39008, Santander, Cantabria, Spain.
| |
Collapse
|
9
|
Murgia A, Veronesi E, Candini O, Caselli A, D’souza N, Rasini V, Giorgini A, Catani F, Iughetti L, Dominici M, Burns JS. Potency Biomarker Signature Genes from Multiparametric Osteogenesis Assays: Will cGMP Human Bone Marrow Mesenchymal Stromal Cells Make Bone? PLoS One 2016; 11:e0163629. [PMID: 27711115 PMCID: PMC5053614 DOI: 10.1371/journal.pone.0163629] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 09/12/2016] [Indexed: 01/15/2023] Open
Abstract
In skeletal regeneration approaches using human bone marrow derived mesenchymal stromal cells (hBM-MSC), functional evaluation before implantation has traditionally used biomarkers identified using fetal bovine serum-based osteogenic induction media and time courses of at least two weeks. However, emerging pre-clinical evidence indicates donor-dependent discrepancies between these ex vivo measurements and the ability to form bone, calling for improved tests. Therefore, we adopted a multiparametric approach aiming to generate an osteogenic potency assay with improved correlation. hBM-MSC populations from six donors, each expanded under clinical-grade (cGMP) conditions, showed heterogeneity for ex vivo growth response, mineralization and bone-forming ability in a murine xenograft assay. A subset of literature-based biomarker genes was reproducibly upregulated to a significant extent across all populations as cells responded to two different osteogenic induction media. These 12 biomarkers were also measurable in a one-week assay, befitting clinical cell expansion time frames and cGMP growth conditions. They were selected for further challenge using a combinatorial approach aimed at determining ex vivo and in vivo consistency. We identified five globally relevant osteogenic signature genes, notably TGF-ß1 pathway interactors; ALPL, COL1A2, DCN, ELN and RUNX2. Used in agglomerative cluster analysis, they correctly grouped the bone-forming cell populations as distinct. Although donor #6 cells were correlation slope outliers, they contrastingly formed bone without showing ex vivo mineralization. Mathematical expression level normalization of the most discrepantly upregulated signature gene COL1A2, sufficed to cluster donor #6 with the bone-forming classification. Moreover, attenuating factors causing genuine COL1A2 gene down-regulation, restored ex vivo mineralization. This suggested that the signature gene had an osteogenically influential role; nonetheless no single biomarker was fully deterministic whereas all five signature genes together led to accurate cluster analysis. We show proof of principle for an osteogenic potency assay providing early characterization of primary cGMP-hBM-MSC cultures according to their donor-specific bone-forming potential.
Collapse
Affiliation(s)
- Alba Murgia
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
| | - Elena Veronesi
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
- TPM, Science & Technology Park for Medicine, Mirandola, Modena, Italia
| | - Olivia Candini
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
| | - Anna Caselli
- CVBF - Consorzio per le Valutazioni Biologiche e Farmacologiche, Ospedale Pediatrico Giovanni XXIII, Bari, Italia
| | - Naomi D’souza
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
| | - Valeria Rasini
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
| | - Andrea Giorgini
- Department of Orthopedic Surgery, University Hospital of Modena and Reggio Emilia, Modena, Italia
| | - Fabio Catani
- Department of Orthopedic Surgery, University Hospital of Modena and Reggio Emilia, Modena, Italia
| | - Lorenzo Iughetti
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
| | - Massimo Dominici
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
- TPM, Science & Technology Park for Medicine, Mirandola, Modena, Italia
- * E-mail: (MD); (JSB)
| | - Jorge S. Burns
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
- TPM, Science & Technology Park for Medicine, Mirandola, Modena, Italia
- * E-mail: (MD); (JSB)
| |
Collapse
|
10
|
Itoi T, Harada Y, Irie H, Sakamoto M, Tamura K, Yogo T, Soeta S, Amasaki H, Hara Y, Tagawa M. Escherichia coli-derived recombinant human bone morphogenetic protein-2 combined with bone marrow-derived mesenchymal stromal cells improves bone regeneration in canine segmental ulnar defects. BMC Vet Res 2016; 12:201. [PMID: 27619812 PMCID: PMC5020464 DOI: 10.1186/s12917-016-0829-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 09/06/2016] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Large bone defects in canines usually require assistance to achieve healing. Implantation of osteoinductive factors can promote bone healing, while transplantation of osteoprogenitor cells can enhance bone regeneration. We hypothesized that implantation of an osteoinductive factor, recombinant human bone morphogenetic protein-2 (rhBMP-2), combined with osteoprogenitor cells, bone marrow-derived mesenchymal stromal cells (BMSCs), would synergistically promote bone healing. In this study, we examined the combined effects of Escherichia coli-derived rhBMP-2 and BMSCs on bone healing after implantation into canine ulnar defects. RESULTS Critical-sized osteoperiosteal segmental defects (2.5 cm) were created in the ulnae of healthy female beagle dogs, and implanted with combinations of E. coli-derived rhBMP-2 (560 or 140 μg) and autologous BMSCs (10(7), 10(5), or 0 cells). In the present study,18 forelimbs of nine healthy purpose-bred female beagles were used. All six treatment groups contained three forelimbs, and the animals were euthanized after 12 weeks. The control groups (560 and 140 μg/0 cells) were cited from our previous study to reduce the number of experimental animals. Radiographically, the regenerated bone width was significantly increased in the 560 or 140 μg with 10(7) and 10(5) cells groups compared with the 0 cells groups. By quantitative CT, the bone mineral density was higher in the 560 μg with 10(7) and 10(5) cells groups, while non-uniformity of the bone mineral density was improved in the 560 μg with 10(7) and 10(5) cells groups and 140 μg/10(7) cells group. Mechanically, the maximum loads at failure were significantly higher in the 560 μg with 10(7) and 10(5) cells groups. Histologically, the regenerated bone was well-developed and contained osteocyte-like cells marrow cavities, and vessels. However, the osteoclasts and osteoblasts were hardly observed. The osteocyte-like cell numbers were significantly higher in the 560 μg with 10(7) and 10(5) cells and 140 μg with 10(7) and 10(5) cells groups. CONCLUSIONS Implantation of E. coli-derived rhBMP-2 and BMSCs led to significantly enhanced bone formation, with improved bone mineral density and reduced non-uniformity of the regenerated bone. Combined implantation of rhBMP-2 and BMSCs may be useful for promotion of bone healing in critical-sized defects in canines.
Collapse
Affiliation(s)
- Takamasa Itoi
- Division of Veterinary Surgery, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino, Tokyo, 180-8602, Japan.
| | - Yasuji Harada
- Division of Veterinary Surgery, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino, Tokyo, 180-8602, Japan
| | - Hiroyuki Irie
- HOYA Technosurgical Corporation, 1-1-110 Tsutsujigaoka, Akishima, Tokyo, 196-0012, Japan
| | - Michiko Sakamoto
- HOYA Technosurgical Corporation, 1-1-110 Tsutsujigaoka, Akishima, Tokyo, 196-0012, Japan
| | - Katsutoshi Tamura
- Division of Animal and Clinical Regenerative Medicine, Kurashiki University of Science and Arts, 2640 Nishinoura, Tsurajima-machi, Kurashiki, Okayama, 712-8505, Japan
| | - Takuya Yogo
- Division of Veterinary Surgery, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino, Tokyo, 180-8602, Japan
| | - Satoshi Soeta
- Division of Veterinary Anatomy, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino, Tokyo, 180-8602, Japan
| | - Hajime Amasaki
- Division of Veterinary Anatomy, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino, Tokyo, 180-8602, Japan
| | - Yasushi Hara
- Division of Veterinary Surgery, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino, Tokyo, 180-8602, Japan
| | - Masahiro Tagawa
- Division of Veterinary Surgery, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino, Tokyo, 180-8602, Japan
| |
Collapse
|
11
|
Smeets R, Barbeck M, Hanken H, Fischer H, Lindner M, Heiland M, Wöltje M, Ghanaati S, Kolk A. Selective laser-melted fully biodegradable scaffold composed of poly(d
,l
-lactide) and β-tricalcium phosphate with potential as a biodegradable implant for complex maxillofacial reconstruction: In vitro
and in vivo
results. J Biomed Mater Res B Appl Biomater 2016; 105:1216-1231. [DOI: 10.1002/jbm.b.33660] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 02/12/2016] [Indexed: 11/08/2022]
Affiliation(s)
- Ralf Smeets
- Department of Oral and Maxillofacial Surgery; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Mike Barbeck
- Frankfurt Orofacial Regenerative Medicine (FORM) Lab, Department for Oral, Cranio-Maxillofacial and Facial Plastic Surgery; Medical Center of the Goethe University Frankfurt; Frankfurt Germany
| | - Henning Hanken
- Department of Oral and Maxillofacial Surgery; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Horst Fischer
- Department of Dental Materials and Biomaterials Research; University Hospital RWTH Aachen; Aachen Germany
| | - Markus Lindner
- Department of Dental Materials and Biomaterials Research; University Hospital RWTH Aachen; Aachen Germany
| | - Max Heiland
- Department of Oral and Maxillofacial Surgery; University Medical Center Hamburg-Eppendorf; Hamburg Germany
| | - Michael Wöltje
- Institute of Textile Machinery and High Performance Material Technology, TU Dresden; Dresden Germany
| | - Shahram Ghanaati
- Frankfurt Orofacial Regenerative Medicine (FORM) Lab, Department for Oral, Cranio-Maxillofacial and Facial Plastic Surgery; Medical Center of the Goethe University Frankfurt; Frankfurt Germany
| | - Andreas Kolk
- Department of Oral- and Maxillofacial Surgery; Klinikum rechts der Isar der Technischen Universität München; 81675 Munich Germany
| |
Collapse
|
12
|
Sworder BJ, Yoshizawa S, Mishra PJ, Cherman N, Kuznetsov SA, Merlino G, Balakumaran A, Robey PG. Molecular profile of clonal strains of human skeletal stem/progenitor cells with different potencies. Stem Cell Res 2015; 14:297-306. [PMID: 25814434 DOI: 10.1016/j.scr.2015.02.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 01/24/2015] [Accepted: 02/18/2015] [Indexed: 12/19/2022] Open
Abstract
Bone marrow stromal cells (BMSCs, also known as bone marrow-derived mesenchymal stem cells) are fibroblastic reticular cells, a subset of which is composed of multipotent skeletal stem cells (SSCs). SSCs/BMSCs are able to recreate a bone/marrow organ in vivo. To determine differences between clonogenic multipotent SSCs and similarly clonogenic but non-multipotent BMSCs, we established single colony-derived strains (SCDSs, initiated by individual Colony Forming Unit-Fibroblasts) and determined their differentiation capacity by vivo transplantation. In this series of human SCDSs (N=24), 20.8% formed fibrous tissue (F), 66.7% formed bone (B), and 12.5% formed a bone/marrow organ, and thus were multipotent (M). RNA isolated from 12 SCDSs just prior to transplantation was analyzed by microarray. Although highly similar, there was variability from one SCDS to another, and SCDSs did not strictly segregate into the three functional groups (F, B or M) by unsupervised hierarchical clustering. We then compared 3 F-SCDSs to 3 M-SCDSs that did segregate. Genes associated with skeletogenesis, osteoblastogeneis, hematopoiesis, and extracellular matrix were over-represented in M-SCDSs compared with F-SCDSs. These results highlight the heterogeneity of SSCs/BMSCs, even between functionally similar SCDSs, but also indicate that differences can be detected that may shed light on the character of the SSC.
Collapse
Affiliation(s)
- Brian J Sworder
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA; Graduate Program in Molecular Medicine, Boston University School of Medicine, Boston, MA 02215, USA
| | - Sayuri Yoshizawa
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Prasun J Mishra
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Natasha Cherman
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Sergei A Kuznetsov
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Glenn Merlino
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Arun Balakumaran
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Pamela G Robey
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA.
| |
Collapse
|
13
|
Robey PG, Kuznetsov SA, Ren J, Klein HG, Sabatino M, Stroncek DF. Generation of clinical grade human bone marrow stromal cells for use in bone regeneration. Bone 2015; 70:87-92. [PMID: 25064527 PMCID: PMC4268331 DOI: 10.1016/j.bone.2014.07.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 07/14/2014] [Accepted: 07/15/2014] [Indexed: 11/22/2022]
Abstract
In current orthopaedic practice, there is a need to increase the ability to reconstruct large segments of bone lost due to trauma, resection of tumors and skeletal deformities, or when normal regenerative processes have failed such as in non-unions and avascular necrosis. Bone marrow stromal cells (BMSCs, also known as bone marrow-derived mesenchymal stem cells), when used in conjunction with appropriate carriers, represent a means by which to achieve bone regeneration in such cases. While much has been done at the bench and in pre-clinical studies, moving towards clinical application requires the generation of clinical grade cells. What is described herein is an FDA-approved cell manufacturing procedure for the ex vivo expansion of high quality, biologically active human BMSCs. This article is part of a Special Issue entitled Stem Cells and Bone.
Collapse
Affiliation(s)
- Pamela G Robey
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA.
| | - Sergei A Kuznetsov
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Jiaqiang Ren
- Department of Transfusion Medicine, Warren G. Magnuson Clinical Center, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA; Cell Processing Section, Department of Transfusion Medicine, Warren G. Magnuson Clinical Center, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Harvey G Klein
- Department of Transfusion Medicine, Warren G. Magnuson Clinical Center, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Marianna Sabatino
- Department of Transfusion Medicine, Warren G. Magnuson Clinical Center, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA; Cell Processing Section, Department of Transfusion Medicine, Warren G. Magnuson Clinical Center, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - David F Stroncek
- Department of Transfusion Medicine, Warren G. Magnuson Clinical Center, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA; Cell Processing Section, Department of Transfusion Medicine, Warren G. Magnuson Clinical Center, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| |
Collapse
|
14
|
Abarrategi A, Perez-Tavarez R, Rodriguez-Milla MA, Cubillo I, Mulero F, Alfranca A, Lopez-Lacomba JL, García-Castro J. In vivo ectopic implantation model to assess human mesenchymal progenitor cell potential. Stem Cell Rev Rep 2014; 9:833-46. [PMID: 23934266 PMCID: PMC3834175 DOI: 10.1007/s12015-013-9464-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Clinical interest on human mesenchymal progenitor cells (hMPC) relies on their potential applicability in cell-based therapies. An in vitro characterization is usually performed in order to define MPC potency. However, in vitro predictions not always correlate with in vivo results and thus there is no consensus in how to really assess cell potency. Our goal was to provide an in vivo testing method to define cell behavior before therapeutic usage, especially for bone tissue engineering applications. In this context, we wondered whether bone marrow stromal cells (hBMSC) would proceed in an osteogenic microenvironment. Based on previous approaches, we developed a fibrin/ceramic/BMP-2/hBMSCs compound. We implanted the compound during only 2 weeks in NOD-SCID mice, either orthotopically to assess its osteoinductive property or subcutaneously to analyze its adequacy as a cell potency testing method. Using fluorescent cell labeling and immunohistochemistry techniques, we could ascertain cell differentiation to bone, bone marrow, cartilage, adipocyte and fibrous tissue. We observed differences in cell potential among different batches of hBMSCs, which did not strictly correlate with in vitro analyses. Our data indicate that the method we have developed is reliable, rapid and reproducible to define cell potency, and may be useful for testing cells destined to bone tissue engineering purposes. Additionally, results obtained with hMPCs from other sources indicate that our method is suitable for testing any potentially implantable mesenchymal cell. Finally, we propose that this model could successfully be employed for bone marrow niche and bone tumor studies.
Collapse
Affiliation(s)
- Ander Abarrategi
- Unidad de Biotecnología Celular, Instituto de Investigación en Enfermedades Raras, Instituto de Salud Carlos III, Carretera Majadahonda-Pozuelo km. 2.200, Majadahonda, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Monfoulet LE, Becquart P, Marchat D, Vandamme K, Bourguignon M, Pacard E, Viateau V, Petite H, Logeart-Avramoglou D. The pH in the microenvironment of human mesenchymal stem cells is a critical factor for optimal osteogenesis in tissue-engineered constructs. Tissue Eng Part A 2014; 20:1827-40. [PMID: 24447025 DOI: 10.1089/ten.tea.2013.0500] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The present study aimed at elucidating the effect of local pH in the extracellular microenvironment of tissue-engineered (TE) constructs on bone cell functions pertinent to new tissue formation. To this aim, we evaluated the osteogenicity process associated with bone constructs prepared from human Bone marrow-derived mesenchymal stem cells (hBMSC) combined with 45S5 bioactive glass (BG), a material that induces alkalinization of the external medium. The pH measured in cell-containing BG constructs was around 8.0, that is, 0.5 U more alkaline than that in two other cell-containing materials (hydroxyapatite/tricalcium phosphate [HA/TCP] and coral) constructs tested. When implanted ectopically in mice, there was no de novo bone tissue in the BG cell-containing constructs, in contrast to results obtained with either HA/TCP or coral ceramics, which consistently promoted the formation of ectopic bone. In addition, the implanted 50:50 composites of both HA/TCP:BG and coral:BG constructs, which displayed a pH of around 7.8, promoted 20-30-fold less amount of bone tissue. Interestingly, hBMSC viability in BG constructs was not affected compared with the other two types of material constructs tested both in vitro and in vivo. Osteogenic differentiation (specifically, the alkaline phosphatase [ALP] activity and gene expression of RUNX2, ALP, and BSP) was not affected when hBMSC were maintained in moderate alkaline pH (≤7.90) external milieu in vitro, but was dramatically inhibited at higher pH values. The formation of mineralized nodules in the extracellular matrix of hBMSC was fully inhibited at alkaline (>7.54) pH values. Most importantly, there is a pH range (specifically, 7.9-8.27) at which hBMSC proliferation was not affected, but the osteogenic differentiation of these cells was inhibited. Altogether, these findings provided evidence that excessive alkalinization in the microenvironment of TE constructs (resulting, for example, from material degradation) affects adversely the osteogenic differentiation of osteoprogenitor cells.
Collapse
Affiliation(s)
- Laurent-Emmanuel Monfoulet
- 1 Laboratory of Bioengineering and Bioimaging for Osteo-Articular Tissues, UMR 7052 CNRS, Université Paris Diderot , Sorbonne Paris Cité, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Cheng N, Dai J, Cheng X, Li S, Miron RJ, Wu T, Chen W, Zhang Y, Shi B. Porous CaP/silk composite scaffolds to repair femur defects in an osteoporotic model. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2013; 24:1963-1975. [PMID: 23674058 PMCID: PMC5995474 DOI: 10.1007/s10856-013-4945-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 04/29/2013] [Indexed: 06/02/2023]
Abstract
The most common complication for patients with postmenopausal osteoporosis is bone-related defects and fractures. While routine medication has a high probability of undesirable side effects, new approaches have aimed to develop regeneration procedures that stimulate new bone formation while reversing bone loss. Recently, we have synthesized a new hybrid CaP/silk scaffold with a CaP-phase distribution and pore architecture better suited to facilitate cell differentiation and bone formation. The aim of the present study was to compare the involved remodeling process and therapeutic effect of porous CaP/silk composite scaffolds upon local implantation into osteoporotic defects. Wistar rats were used to induce postmenopausal osteoporotic model by bilateral ovariectomy. The pure silk and hybrid CaP/silk scaffolds were implanted into critical sized defects created in distal femoral epiphysis. After 14 and 28 days, the in vivo osteogenetic efficiency was evaluated by μCT analysis, hematoxylin and eosin staining, Safranin O staining, tartrate-resistant acid phosphatase staining, and immunohistochemical assessment. Animals with or without critical-sized defects were used as drill or blank controls, respectively. The osteoporotic defect model was well established with significantly decreased μCT parameters of BV/TV, Tb.N and increased Tb.Sp, porosity, combined with changes in histological observations. During the healing process, the critical-sized drill control defects failed to regenerate appreciable bone tissue, while more significantly increased bone formation and mineralization with dynamic scaffold degradation and decreased osteoclastic bone resorption could be detected within defects with hybrid CaP/silk scaffolds compared to pure silk scaffolds.
Collapse
Affiliation(s)
- Ning Cheng
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Zhang Y, Cheng N, Miron R, Shi B, Cheng X. Delivery of PDGF-B and BMP-7 by mesoporous bioglass/silk fibrin scaffolds for the repair of osteoporotic defects. Biomaterials 2012; 33:6698-708. [PMID: 22763224 DOI: 10.1016/j.biomaterials.2012.06.021] [Citation(s) in RCA: 135] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Accepted: 06/15/2012] [Indexed: 01/23/2023]
Abstract
Osteoporosis is a chronic disease affecting millions of people worldwide caused by an imbalance between bone-forming osteoblasts and bone-resorbing osteoclasts. Despite recent developments in pharmacological agents to prevent osteoporotic-related fractures, much less attention has been placed on the repair of bone defects following fracture. Critical to this process is the recruitment of mesenchymal stem cells (MSCs) to defect sites by growth factors. One method which has been effective for the sustained release of growth factors is that of gene therapy. The aim of the present study was to investigate newly developed mesoporous bioglass/silk fibrin scaffolds containing adPDGF-b and adBMP-7 into osteoporotic critical-sized femur defects in ovariectomised rats following treatment periods of 2 and 4 weeks. In vivo osteogenetic efficiency evaluated by μ-CT analysis, hematoxylin and eosin staining, and immunohistochemical (type I collagen, osteopontin and BSP) revealed significantly new bone formation in defects containing adenovirus for both PDGF-b and BMP-7 when compared to scaffolds alone and scaffolds containing BMP-7. TRAP-positive staining also demonstrated the ability for these scaffolds to be degraded over time and initiate bone turnover/remodeling. Although the use of gene therapy for clinical applications is still in its infancy, results from the present study demonstrate their potent ability to recruit mesenchymal progenitor cells through sustained release of PDGF-b and BMP-7 which may be beneficial for patients suffering from osteoporotic-related fractures.
Collapse
Affiliation(s)
- Yufeng Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan 430079, PR China.
| | | | | | | | | |
Collapse
|
18
|
Agata H, Yamazaki M, Uehara M, Hori A, Sumita Y, Tojo A, Kagami H. Characteristic differences among osteogenic cell populations of rat bone marrow stromal cells isolated from untreated, hemolyzed or Ficoll-treated marrow. Cytotherapy 2012; 14:791-801. [PMID: 22494074 DOI: 10.3109/14653249.2012.674639] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AIMS Although bone marrow (BM) stromal cells (SC; BMSC) isolated from adherent cultures of untreated BM are known to contain both committed and uncommitted osteogenic cells, it remains unknown whether BMSC isolated either by hemolysis or Ficoll centrifugation also contain both of these populations. METHODS Differences in the osteogenic cell populations of rat BMSC isolated from untreated, hemolyzed or Ficoll-treated BM were analyzed by in vivo transplantation, flow cytometry, alkaline phosphatase (ALP) assay, real-time polymerase chain reaction (PCR) and alizarin red staining. RESULTS Transplantation of non-cultured samples indicated that the Ficolled BMSC contained the lowest number of committed osteogenic cells. Flow cytometric analysis of cultured, non-induced samples showed that the percentage of ALP-positive cells was significantly lower in Ficolled BMSC. Quantitative ALP assays confirmed that the lowest ALP activity was in the Ficolled BMSC. Hemolyzed BMSC also contained lower numbers of committed osteogenic cells than untreated BMSC, but still more than Ficolled BMSC. Interestingly, the Ficolled BMSC showed the greatest levels of osteogenic ability when cultured in osteogenic induction medium. CONCLUSIONS These findings suggest that, although Ficolled BMSC rarely contain committed osteogenic cells, they are able to show comparable or even greater levels of osteogenic ability after induction, possibly because they contain a greater proportion of uncommitted stem cells. In contrast, induction is optional but recommended for both untreated and hemolyzed BMSC before use, because both these groups contain both committed and uncommitted osteogenic cells. These findings are of significant importance when isolating BMSC for use in bone tissue engineering.
Collapse
Affiliation(s)
- Hideki Agata
- Tissue Engineering Research Group, Division of Molecular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan.
| | | | | | | | | | | | | |
Collapse
|
19
|
Lomelino RDO, Castro-Silva II, Linhares ABR, Alves GG, Santos SRDA, Gameiro VS, Rossi AM, Granjeiro JM. The association of human primary bone cells with biphasic calcium phosphate (βTCP/HA 70:30) granules increases bone repair. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2012; 23:781-788. [PMID: 22201029 DOI: 10.1007/s10856-011-4530-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Accepted: 12/09/2011] [Indexed: 05/31/2023]
Abstract
This work evaluates the suitability of biphasic calcium phosphate (BCP) granules (β-TCP/HA 70:30) as potential carriers for cell-guided bone therapy. The BCP granules were obtained by synthesis in the presence of wax, thermal treatment, crushing and sieving and characterized by scanning electron microscopy (SEM), X-ray diffraction and Fourier transform infrared spectroscopy. The cytocompatibility of the BCP granules was confirmed by a multiparametric cytotoxicity assay. SEM analysis showed human bone cell adhesion and migration after seeding onto the material. Rat subcutaneous xenogeneic grafting of granules associated to human bone cells revealed a more accentuated moderate chronic inflammatory infiltrate, without signs of a strong xenoreactivity. Histomorphometrical analysis of bone repair of defects in rat skulls (∅ = 5 mm) has shown that bone cell associated-BCP and autograft promoted a two- and threefold increase, respectively, on new bone formation after 45 days, as compared to BCP alone and blood clot. The increase in bone repair supports the suitability the biocompatible (70:30) BCP granules as injectable and mouldable scaffolds for human cells in bone bioengineering.
Collapse
|
20
|
Mankani MH, Afghani S, Franco J, Launey M, Marshall S, Marshall GW, Nissenson R, Lee J, Tomsia AP, Saiz E. Lamellar spacing in cuboid hydroxyapatite scaffolds regulates bone formation by human bone marrow stromal cells. Tissue Eng Part A 2011; 17:1615-23. [PMID: 21294634 DOI: 10.1089/ten.tea.2010.0573] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND A major goal in bone engineering is the creation of large volume constructs (scaffolds and stem cells) that bear load. The scaffolds must satisfy two competing requirements--they need be sufficiently porous to allow nutrient flow to maintain cell viability, yet sufficiently dense to bear load. We studied the effect of scaffold macroporosity on bone formation and scaffold strength, for bone formed by human bone marrow stromal cells. METHODS Rigid cubical hydroxyapatite/tricalcium phosphate scaffolds were produced by robo-casting. The ceramic line thickness was held constant, but the distance between adjacent lines was either 50, 100, 200, 500, or 1000 μm. Cultured human bone marrow stromal cells were combined with the scaffolds in vitro; transplants were placed into the subcutis of immunodeficient mice. Transplants were harvested 9, 18, 23, 38, or 50 weeks later. Bone formation and scaffold strength were analyzed using histology and compression testing. RESULTS Sixty transplants were evaluated. Cortical bone increased with transplant age, and was greatest among 500 μm transplants. In contrast, maximum transplant strength was greatest among 200 μm transplants. CONCLUSIONS Lamellar spacing within scaffolds regulates the extent of bone formation; 500 μm yields the most new bone, whereas 200 μm yields the strongest transplants.
Collapse
Affiliation(s)
- Mahesh H Mankani
- Department of Surgery, University of California-San Francisco, San Francisco, California 94143-0807, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Taichman RS, Wang Z, Shiozawa Y, Jung Y, Song J, Balduino A, Wang J, Patel LR, Havens AM, Kucia M, Ratajczak MZ, Krebsbach PH. Prospective identification and skeletal localization of cells capable of multilineage differentiation in vivo. Stem Cells Dev 2011; 19:1557-70. [PMID: 20446812 DOI: 10.1089/scd.2009.0445] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
A prospective in vivo assay was used to identify cells with potential for multiple lineage differentiation. With this assay, it was first determined that the 5-fluorouracil resistant cells capable of osseous tissue formation in vivo also migrated toward stromal derived factor-1 (SDF-1) in vitro. In parallel, an isolation method based on fluorescence-activated cell sorting was employed to identify a very small cell embryonic-like Lin-/Sca-1+CD45- cell that with as few as 500 cells was capable of forming bone-like structures in vivo. Differential marrow fractionation studies determined that the majority of the Lin-Sca-1+CD45- cells reside in the subendosteal regions of marrow. To determine whether these cells were capable of differentiating into multiple lineages, stromal cells harvested from Col2.3 Delta TK mice were implanted with a gelatin sponge into SCID mice to generate thymidine kinase sensitive ossicles. At 1.5 months, 2,000 green fluorescent protein (GFP)+ Lin-Sca-1+CD45- cells were injected into the ossicles. At harvest, colocalization of GFP-expressing cells with antibodies to the osteoblast-specific marker Runx-2 and the adipocyte marker PPAP gamma were observed. Based on the ability of the noncultured cells to differentiate into multiple mesenchymal lineages in vivo and the ability to generate osseous tissues at low density, we propose that this population fulfills many of the characteristics of mesenchymal stem cells.
Collapse
Affiliation(s)
- Russell S Taichman
- Department of Periodontics and Oral Medicine, University of Michigan, Ann Arbor, Michigan 48109-1078, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Bacci C, Lucchiari N, Valente M, Della Barbera M, Frigo AC, Berengo M. Intra-oral bone harvesting: two methods compared using histological and histomorphometric assessments. Clin Oral Implants Res 2010; 22:600-5. [PMID: 21070379 DOI: 10.1111/j.1600-0501.2010.02022.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
AIM This study used morphometric analyses to compare two methods for the intra-oral harvesting of particulate bone: Mectron Piezosurgery® and the Meta Micross®. MATERIALS AND METHODS Twenty patients requiring bilateral germectomy of the lower third molars for orthodontic reasons were selected and a sample was harvested from each patient from a standardised donor site (the cortical bone in the area of the retromolar triangle). Ten samples were obtained for each method. The particulate collected were subjected to a histological examination and the samples were analysed considering the following parameters: the mean surface area of fragments, the mean surface area considered vital and the mean surface area considered non-vital, the mean percentage of area considered vital and the mean percentage of area considered non-vital, the mean number of normal osteocytes and the mean number of osteocytes with morphological changes identified per unit area (600,000 μm(2)). The results were analysed, calculating the mean and the corresponding standard deviations, and testing their significance using Student's t-test, and plotted in graphs. RESULTS Mectron Piezosurgery® produced significantly larger particles (P<0.05) than the Meta Micross®, with a larger mean surface area considered vital and a significantly larger (P<0.05) surface area considered non-vital. Mectron Piezosurgery® also produced a smaller mean percentage of area considered vital (64.83%) and a larger mean percentage of area considered non-vital (35.17%) by comparison with the Meta Micross® (75.34% and 24.66%, respectively). The data also showed that the two methods produce a similar quantity of empty lacunae, and that the Mectron Piezosurgery® produces a larger quantity of osteocytes. CONCLUSIONS The analyses conducted demonstrated that the particulate collected with the Meta Micross® had a smaller mean surface area of the fragments and a smaller surface area of bone considered non-vital than in the particulate collected using Mectron Piezosurgery®.
Collapse
Affiliation(s)
- Christian Bacci
- Department of Surgical and Medical Specialities, Oral Surgery-Section of Dentistry, University of Padova, Padova, Italy.
| | | | | | | | | | | |
Collapse
|
23
|
Giannicola G, Ferrari E, Citro G, Sacchetti B, Corsi A, Riminucci M, Cinotti G, Bianco P. Graft vascularization is a critical rate-limiting step in skeletal stem cell-mediated posterolateral spinal fusion. J Tissue Eng Regen Med 2010; 4:273-83. [PMID: 20013838 DOI: 10.1002/term.238] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The ability of skeletal stem cells (SSCs) to direct spinal fusion (SF) upon transplantation in conjunction with osteoconductive biomaterials was investigated in a rabbit model. When tested in a mouse heterotopic transplantation assay, rabbit SSCs and Pro-Osteon 500R was osteoconductive and supported osteogenesis. When used in a SF model, the same constructs induced bone formation in periapophyseal regions (PARs). In this respect, they proved to be superior to grafts of cell-free carrier or total uncultured bone marrow-carrier constructs, used as controls. However, interapophyseal regions (IARs) remained devoid of new bone, such that true bony bridging of adjacent transverse apophyses (true SF) could not be achieved. Interestingly, this could not be predicted from high-resolution radiography. A systematic histological survey of the entire graft harvested at 6 months was essential for proper assessment of the transplantation procedure outcome. Immunohistochemical analysis of microvessel density revealed that IARs remained undervascularized, as compared to PARs, suggesting that differential vascularization could account for the absence or presence of new bone formation in the same regions. SF is an extreme model of stem cell-directed bone regeneration, requiring a combination of orthotopic (PAR) and heterotopic (IAR) bone formation. Our data show that, in this setting, graft size can be critical with respect to the necessary neovascularization, a crucial variable independent of proper osteogenic and osteoconductive competence of the cells and materials employed. Furthermore, stringent histological studies are mandatory for proper assessment of outcomes in SF studies, in which the use of mineralized materials can make radiographic assessment misleading.
Collapse
Affiliation(s)
- Giuseppe Giannicola
- Orthopaedic Department, University 'La Sapienza', Piazzale Aldo Moro 5, 00185 Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Superparamagnetic iron oxide nanoparticles labeling of bone marrow stromal (mesenchymal) cells does not affect their "stemness". PLoS One 2010; 5:e11462. [PMID: 20628641 PMCID: PMC2898800 DOI: 10.1371/journal.pone.0011462] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Accepted: 05/07/2010] [Indexed: 12/17/2022] Open
Abstract
Superparamagnetic iron oxide nanoparticles (SPION) are increasingly used to label human bone marrow stromal cells (BMSCs, also called “mesenchymal stem cells”) to monitor their fate by in vivo MRI, and by histology after Prussian blue (PB) staining. SPION-labeling appears to be safe as assessed by in vitro differentiation of BMSCs, however, we chose to resolve the question of the effect of labeling on maintaining the “stemness” of cells within the BMSC population in vivo. Assays performed include colony forming efficiency, CD146 expression, gene expression profiling, and the “gold standard” of evaluating bone and myelosupportive stroma formation in vivo in immuncompromised recipients. SPION-labeling did not alter these assays. Comparable abundant bone with adjoining host hematopoietic cells were seen in cohorts of mice that were implanted with SPION-labeled or unlabeled BMSCs. PB+ adipocytes were noted, demonstrating their donor origin, as well as PB+ pericytes, indicative of self-renewal of the stem cell in the BMSC population. This study confirms that SPION labeling does not alter the differentiation potential of the subset of stem cells within BMSCs.
Collapse
|
25
|
Kasten P, Beyen I, Bormann D, Luginbühl R, Plöger F, Richter W. The effect of two point mutations in GDF-5 on ectopic bone formation in a beta-tricalciumphosphate scaffold. Biomaterials 2010; 31:3878-84. [PMID: 20170953 DOI: 10.1016/j.biomaterials.2010.01.109] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2009] [Accepted: 01/18/2010] [Indexed: 12/14/2022]
Abstract
The osteoinductivity of human growth-and-differentiation factor-5 (GDF-5) is well established, but a reduced amount of ectopic bone is formed compared to other members of the bone morphogenetic protein (BMP) family like BMP-2. We hypothesized that swap of two BMP-receptor-interacting residues of GDF-5 to amino acids present in BMP-2 (methionine to valine at the sites 453 and 456) may improve the bone formation capacity of the mutant GDF-5. Heterotopic bone formation of a mutant GDF-5 coated beta-TCP carrier was compared to carriers coated with similar amounts (10 microg) of GDF-5 and BMP-2 in SCID mice. Four week explants revealed 6-fold higher ALP activity in the mutant GDF-5 versus the wild type GDF-5 group (p < 0.0001) and 1.4-fold higher levels compared to BMP-2 (p < 0.006). Bone area in histology was significantly higher in mutant GDF-5 versus all other groups at 4 weeks; however, at 8 weeks BMP-2 reached a similar neo-bone formation like mutant GDF-5. Micro-CT evaluation confirmed higher values in the mutant GDF-5 and BMP-2 groups compared to wild type GDF-5. In conclusion, the mutant GDF-5 showed superior bone formation capacity than GDF-5, and a faster induction at similar final outcome as BMP-2. Mutant GDF-5 thus represents a promising new GDF-5 variant for bone regeneration possibly acting via an increased binding affinity to the BMP-type I receptor.
Collapse
Affiliation(s)
- Philip Kasten
- Research Center for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
26
|
Comparison of computed tomography and microradiography for graft evaluation after reconstruction of critical size bone defects using beta-tricalcium phosphate. J Craniomaxillofac Surg 2009; 38:38-46. [PMID: 19700333 DOI: 10.1016/j.jcms.2009.07.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Revised: 05/21/2009] [Accepted: 07/28/2009] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION The aim of the study was to evaluate the accuracy of computed tomography (CT) for in vivo follow up after mandibular reconstruction. MATERIAL AND METHODS Unilateral mandibular defects were surgically created in ten sheep and either reconstructed using blood soaked beta-tricalcium phosphate (beta-TCP) cylinders (group A, n=5) or blood soaked beta-TCP cylinders that were additionally loaded with autologous bone marrow (group B, n=5). The two graft designs resulted in different stages of graft ossification representative of different stages of healing. CT datasets were fused with microradiographs and measurements of ceramic area based on both methods were compared. RESULTS Two animals (groups A (n=1) and B (n=1)) presented infection and graft dislocation that was visible on CT and were excluded from statistical evaluation. Group A grafts underwent moderate degradation (53.55%+/-9.7) and incomplete bony incorporation representing an intermediate state of healing while ceramic grafts within group B developed a high grade of osseointegration and degradation (94.2%+/-3.3) consistent with progressive healing. Statistical comparison of measurements based on both methods revealed a significant bias (p<0.05) and a non-significant variance for group A and a significant variance (p<0.05) and non-significant bias for group B. CONCLUSION Our results indicate that conventional CT is not suitable to objectively evaluate ossification and degradation of a beta-TCP graft in vivo and further attempts to improve clinical visualization of beta-TCP need to be undertaken.
Collapse
|
27
|
Mankani MH, Kuznetsov SA, Marshall GW, Robey PG. Creation of new bone by the percutaneous injection of human bone marrow stromal cell and HA/TCP suspensions. Tissue Eng Part A 2009; 14:1949-58. [PMID: 18800877 DOI: 10.1089/ten.tea.2007.0348] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The in vivo transplantation assay has become a valuable tool for assessing the osteogenic potential of diverse cell populations. It has required that cells are cotransplanted with a matrix into recipient animals using large incisions and extensive dissections. Here, we demonstrate that transplants of an osteogenic cell population, bone marrow stromal cells (BMSCs), are capable of assembling into mature bone organs when injected as suspensions of cells and a particulate matrix. METHODS Human BMSCs, along with hydroxyapatite/tricalcium phosphate (HA/TCP) particles, were placed either into the dorsal subcutaneous space or onto the calvarium of immunodeficient mice, either via injection or via a wide operative exposure. Transplants were harvested from 7 to 110 weeks later; their histologic and mechanical properties and their cellular origin were analyzed. RESULTS A total of 43 transplants were evaluated. The extent of new bone and hematopoiesis, the bone's adherence to the underlying mouse calvarium, and the bone elastic modulus and hardness were comparable between the two groups. In situ hybridization confirmed a human origin of the new bone. CONCLUSIONS Our data indicate that BMSCs and HA/TCP particles, when injected as a suspension, can assemble into mature bone organs, and that this bone has histologic and mechanical properties similar to bone formed in standard transplants delivered through a large incision. These results open the possibility for assessing the osteogenic capacities of cell populations, for modeling bone formation and repair and for treating bone deficits, all in the context of minimal surgical intervention or soft tissue disruption.
Collapse
Affiliation(s)
- Mahesh H Mankani
- Division of Plastic Surgery, Department of Surgery, University of California-San Francisco, San Francisco, California 94143-0807, USA.
| | | | | | | |
Collapse
|
28
|
Microporous nanofibrous fibrin-based scaffolds for bone tissue engineering. Biomaterials 2008; 29:4091-9. [PMID: 18640716 DOI: 10.1016/j.biomaterials.2008.06.030] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Accepted: 06/28/2008] [Indexed: 11/22/2022]
Abstract
The fibrotic response of the body to synthetic polymers limits their success in tissue engineering and other applications. Though porous polymers have demonstrated improved healing, difficulty in controlling their pore sizes and pore interconnections has clouded the understanding of this phenomenon. In this study, a novel method to fabricate natural polymer/calcium phosphate composite scaffolds with tightly controllable pore size, pore interconnection, and calcium phosphate deposition was developed. Microporous, nanofibrous fibrin scaffolds were fabricated using sphere-templating methods. Composite scaffolds were created by solution deposition of calcium phosphate on fibrin surfaces or by direct incorporation of nanocrystalline hydroxyapatite (nHA). The SEM results showed that fibrin scaffolds exhibited a highly porous and interconnected structure. Osteoblast-like cells, obtained from murine calvaria, attached, spread and showed a polygonal morphology on the surface of the biomaterial. Multiple cell layers and fibrillar matrix deposition were observed. Moreover, cells seeded on mineralized fibrin scaffolds exhibited significantly higher alkaline phosphatase activity as well as osteoblast marker gene expression compared to fibrin scaffolds and nHA incorporated fibrin scaffolds (0.25 and 0.5g). All types of scaffolds were degraded both in vitro and in vivo. Furthermore, these scaffolds promoted bone formation in a mouse calvarial defect model and the bone formation was enhanced by addition of rhBMP-2.
Collapse
|
29
|
Applications of an athymic nude mouse model of nonhealing critical-sized calvarial defects. J Craniofac Surg 2008; 19:192-7. [PMID: 18216688 DOI: 10.1097/scs.0b013e31815c93b7] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Calvarial bone defects are a common clinical scenario in craniofacial surgery. Numerous approaches are used to reconstruct skull defects, and each possesses its own inherent disadvantages. This fact underscores the opportunity to develop a novel method to repair osseous defects in craniofacial surgery. Recent literature strongly suggests that cell-based therapies in the form of regenerative medicine may be a developing paradigm in reconstructive surgery. Although numerous studies have probed osteoprogenitor cells from mice, few have explored the biology of human cells in the setting of osteogenesis in an equally rigorous manner. This study proposes a nude mouse model of critical-sized calvarial defects to study the in vivo biology of human osteoprogenitor cells. Critical-sized 4.0-mm calvarial defects were created in nude mice (n = 15) with a custom trephine drill bit outfitted to a dental drill handpiece. During the craniotomy, the dura mater was spared from injury. Gross inspection, routine histology, and micro-computed tomographic scanning were performed at 2, 4, 8, and 16 weeks postoperatively. There was no calvarial healing in any of the animals by 16 weeks. The dura mater remained intact in all subjects. Gross, histologic, and radiographic assays confirmed these findings. Although several studies have implanted human osteoprogenitor cells in vivo in various animal models, few have documented the appropriate controls or conditions necessary to support the potential to translate benchtop findings into clinical applications. We propose in this study that the nude mouse critical-sized calvarial defect model will be valuable with increasing investigations with human osteoprogenitor cells.
Collapse
|
30
|
Hayashi-Sakai S, Sakai J, Kitamura T, Sakamoto M, Taguchi Y. The clinical oro-facial findings of an 11-year-old Japanese boy with 47, XYY: A case report. PEDIATRIC DENTAL JOURNAL 2008. [DOI: 10.1016/s0917-2394(08)70137-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
31
|
Mankani MH, Kuznetsov SA, Robey PG. Formation of hematopoietic territories and bone by transplanted human bone marrow stromal cells requires a critical cell density. Exp Hematol 2007; 35:995-1004. [PMID: 17960668 DOI: 10.1016/j.exphem.2007.01.051] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Bone marrow stromal cells (BMSCs) include multipotent cells with the ability to form mature bone organs upon in vivo transplantation. Hematopoiesis in these bone organs has been ascribed to the action of skeletal stem cells, which are capable of differentiating towards bone and hematopoiesis-supporting stroma. Yet, the creation of hematopoietic territories may be in part a natural consequence of the formation of a sufficiently mature and large bone microenvironment. Here, we describe, for the first time, a relationship between BMSC numbers and the extent of bone/hematopoiesis formation in heterotopic transplants. METHODS Human BMSCs were transplanted along with hydroxyapatite/tricalcium phosphate, utilizing a spectrum of dosages, into immunotolerant mice; the transplants were followed for up to 29 months. RESULTS The extent of bone and hematopoiesis formation increased with increasing BMSC numbers; however, the relationship was sigmoid in character, and a threshold number of BMSCs was necessary for extensive bone formation or any hematopoiesis. Hematopoiesis only occurred in conjunction with extensive bone formation, and no hematopoiesis occurred where bone formation was poor. Consistent with our earlier studies of long-term BMSC transplantation, the transplants underwent a change in bone morphology but not bone content after 8 weeks. CONCLUSION Our results have provided evidence that the formation of both hematopoiesis and a mature bone organ is as much a consequence of a sufficiently high local density of bone marrow stromal cells as it is the product of skeletal stem cell action.
Collapse
Affiliation(s)
- Mahesh H Mankani
- Division of Plastic Surgery, Department of Surgery, University of California-San Francisco, San Francisco, Calif., USA.
| | | | | |
Collapse
|
32
|
Abstract
BACKGROUND Stem cells have been isolated from a variety of embryonic and postnatal (adult) tissues, including bone marrow. Bone marrow stromal cells (BMSCs), which are non-blood-forming cells in marrow, contain a subset of skeletal stem cells (SSCs) that are able to regenerate all types of skeletal tissue: bone, cartilage, blood-supportive stromal cells and marrow fat cells. METHODS Bone marrow suspensions are placed into culture for analysis of their biological character and for expansion of their number. The resulting populations of cells are used in a variety of assays to establish the existence of an adult SSC, and the ability of BMSC populations to regenerate hard tissues in the craniofacial region, in conjunction with appropriate scaffolds. RESULTS Single-cell analysis established the existence of a true adult SSC in bone marrow. Populations of ex vivo expanded BMSCs (a subset of which are SSCs) are able to regenerate a bone/marrow organ. In conjunction with appropriate scaffolds, these cells can be used to regenerate bone in a variety of applications. CONCLUSIONS BMSCs have the potential to re-create tissues of the craniofacial region to restore normal structure and function in reconstructing the hard tissues of a face. Ex vivo expanded BMSCs with scaffolds have been used in a limited number of patients to date, but likely will be used more extensively in the near future.
Collapse
Affiliation(s)
- Pamela Gehron Robey
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA.
| | | |
Collapse
|
33
|
Mankani MH, Kuznetsov SA, Wolfe RM, Marshall GW, Robey PG. In vivo bone formation by human bone marrow stromal cells: reconstruction of the mouse calvarium and mandible. Stem Cells 2006; 24:2140-9. [PMID: 16763200 DOI: 10.1634/stemcells.2005-0567] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Bone marrow stromal cells (BMSCs) contain a subset of multipotent cells with the potential to repair hard-tissue defects. Mouse BMSCs, combined with a collagen carrier, can close critical-sized homologous mouse calvarial defects, but this new bone has a poor union with the adjacent calvarium. When human BMSCs are transplanted for the purpose of engineering new bone, best results can be achieved if the cells are combined with hydroxyapatite/tricalcium phosphate (HA/TCP) particles. Here, we demonstrate that transplantation of cultured human BMSCs in conjunction with HA/TCP particles can be used successfully to close mouse craniofacial bone defects and that removal of the periosteum from the calvarium significantly enhances union with the transplant. Transplants were followed for up to 96 weeks and were found to change in morphology but not bone content after 8 weeks; this constitutes the first description of human BMSCs placed long-term to heal bone defects. New bone formation continued to occur in the oldest transplants, confirmed by tetracycline labeling. Additionally, the elastic modulus of this engineered bone resembled that of the normal mouse calvarium, and our use of atomic force microscopy (AFM)-based nanoindentation offered us the first opportunity to compare these small transplants against equally minute mouse bones. Our results provide insights into the long-term behavior of newly engineered orthotopic bone from human cells and have powerful implications for therapeutic human BMSC transplantation.
Collapse
Affiliation(s)
- Mahesh H Mankani
- Department of Surgery, Division of Plastic Surgery, University of California, San Francisco, 1001 Potrero Avenue, Box 0807, 94143-0807, USA.
| | | | | | | | | |
Collapse
|
34
|
Mankani MH, Kuznetsov SA, Shannon B, Nalla RK, Ritchie RO, Qin Y, Robey PG. Canine cranial reconstruction using autologous bone marrow stromal cells. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 168:542-50. [PMID: 16436668 PMCID: PMC1606510 DOI: 10.2353/ajpath.2006.050407] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Limited-sized transplants of culture-expanded autologous or allogeneic bone marrow stromal cells (BMSCs) form cortico-cancellous bone in rodent models. Initiation of clinical studies using autologous BMSC transplantation requires effective bone formation among sizable transplants in a large animal model as well as noninvasive techniques for evaluating transplant success. Here, we obtained bone marrow from the femurs of six dogs and expanded BMSCs in tissue culture. Autologous BMSC-hydroxyapatite/tricalcium phosphate (HA/TCP) transplants were introduced into critical-sized calvarial defects and contralateral control skull defects received HA/TCP vehicle alone. At intervals ranging from 2 to 20 months, transplants were biopsied or harvested for histological and mechanical analysis. Noninvasive studies, including quantitative computed tomography scans and ultrasound, were simultaneously obtained. In all animals, BMSC-containing transplants formed significantly more bone than their control counterparts. BMSC-associated bone possessed mechanical properties similar to the adjacent normal bone, confirmed by both ultrasound and ex vivo analysis. Evaluation by quantitative computed tomography confirmed that the extent of bone formation demonstrated by histology could be discerned through noninvasive means. These results show that autologous cultured BMSC transplantation is a feasible therapy in clinical-sized bone defects and that such transplants can be assessed noninvasively, suggesting that this technique has potential for use in patients with certain bone defects.
Collapse
Affiliation(s)
- Mahesh H Mankani
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143-0807, USA.
| | | | | | | | | | | | | |
Collapse
|
35
|
Eliopoulos N, Stagg J, Lejeune L, Pommey S, Galipeau J. Allogeneic marrow stromal cells are immune rejected by MHC class I- and class II-mismatched recipient mice. Blood 2005; 106:4057-65. [PMID: 16118325 DOI: 10.1182/blood-2005-03-1004] [Citation(s) in RCA: 418] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
It has been suggested that marrow stromal cells (MSCs) may be immunoprivileged and can engraft in allogeneic recipients with intact immune systems. We determined if the implantation of murine MSCs engineered to release erythropoietin (Epo) would be feasible in major histocompatibility complex (MHC)-mismatched allogeneic mice without immunosuppression, and we monitored hematocrit (Hct) as a reporter of MSC graft survival. MSCs from C57Bl/6 mice were engineered to release murine Epo (Epo+ MSCs) and implanted subcutaneously in either syngeneic C57Bl/6 mice or MHC-mismatched Balb/c mice. In syngeneic recipients, the Hct rapidly rose from baseline level and remained higher than .88 (88%) for more than 200 days. However, in MHC-mismatched recipient Balb/c mice, the Hct rose transiently and rapidly declined to baseline values. Repeat implantations in these same mice were associated with an acquired refractoriness in the Hct response consistent with alloimmunization to donor Epo+ MSCs. Allogeneic MSC implants had an increased proportion of host-derived lymphoid CD8+, natural killer T (NKT), and NK infiltrating cells compared with syngeneic controls, and splenocytes isolated from Balb/c mice that had received implants also displayed a significant interferon-gamma (IFNgamma) response to C57Bl/6 MSCs in vitro. These results strongly suggest that MSCs are not intrinsically immunoprivileged and cannot serve as a "universal donor" in immunocompetent MHC-mismatched recipients.
Collapse
Affiliation(s)
- Nicoletta Eliopoulos
- Lady Davis Institute for Medical Research, McGill University, 3755 Cote St-Catherine Rd, Montreal, QC Canada H3T1E2
| | | | | | | | | |
Collapse
|
36
|
Abstract
STUDY DESIGN Retrospective study of 20 patients with degenerative cervical spinal canal stenosis who were treated with corpectomy followed by the placement of a distractable titanium cage (anterior distraction device [ADD]) (Ulrich GmbH & Co. KG, Ulm, Germany). OBJECTIVE To investigate the efficacy of distractable titanium cages for reconstruction following decompressive cervical corpectomy. SUMMARY OF BACKGROUND DATA Anterior corpectomy and reconstruction for extensive spinal canal stenosis are standard techniques. The reconstruction is commonly performed with bone graft and anterior plating. Alternatively, different fusion cages have gained acceptance in spine surgery. However, in traditionally designed cages, an over-distraction of the involved segment is necessary to place the cage into the defect, and to achieve tight contact between cage and endplates. The new designed cage (ADD) can be distracted in situ. Therefore, the height of the cage can be adjusted precisely to the size of the corpectomy. METHODS All patients were treated with corpectomy followed by the placement of an ADD. A cervical plate was added in 13 cases, and an "All-in-one-system" (i.e., a cage with an attached plate with 2 holes for screw fixation on both sides, ADD) was used in 7. Follow-up was 18-60 months. Clinical evaluation and radiographs were performed after 3, 6, and 12 months, and then once a year. RESULTS In all cases, a stable reconstruction of the anterior column was achieved, with no signs of instability on flexion/extension radiographs. However, there was one incidence of cage subsidence. According to Odom criteria, 11 patients (55%) had excellent outcome, 4 (20%) good, 3 (15%) satisfactory, and 2 (10%) poor. CONCLUSIONS The results of the study show that distractable cages are useful vertebral body replacements because they can be adjust to the size of the corpectomy in situ and provide immediate strong anterior column support, avoiding bone graft site morbidity. The possibility of direct cage fixation without an additional plate simplifies the procedure.
Collapse
Affiliation(s)
- Christian Woiciechowsky
- Department of Neurosurgery, Charité, Campus Virchow-Klinkum, University Medicine of Berlin, Berlin, Germany.
| |
Collapse
|
37
|
Eghbali-Fatourechi GZ, Lamsam J, Fraser D, Nagel D, Riggs BL, Khosla S. Circulating osteoblast-lineage cells in humans. N Engl J Med 2005; 352:1959-66. [PMID: 15888696 DOI: 10.1056/nejmoa044264] [Citation(s) in RCA: 297] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND Although current evidence suggests that only a minuscule number of osteoblast-lineage cells are present in peripheral blood, we hypothesized that such cells circulate but that their concentration has been vastly underestimated owing to the use of assays that required adherence to plastic. We further reasoned that the concentration of these cells is elevated during times of increased bone formation, such as during pubertal growth. METHODS We used flow cytometry with antibodies to bone-specific proteins to identify circulating osteoblast-lineage cells in 11 adolescent males and 11 adult males (mean [+/-SD] age, 14.5+/-0.7 vs. 37.7+/-7.6 years). Gene expression and in vitro and in vivo bone-forming assays were used to establish the osteoblastic lineage of sorted cells. RESULTS Cells positive for osteocalcin and cells positive for bone-specific alkaline phosphatase were detected in the peripheral blood of adult subjects (1 to 2 percent of mononuclear cells). There were more than five times as many cells positive for osteocalcin in the circulation of adolescent boys (whose markers of bone formation were clearly increased as a result of pubertal growth) as compared with adult subjects (P<0.001). The percentage of cells positive for osteocalcin correlated with markers of bone formation. Sorted osteocalcin-positive cells expressed osteoblastic genes, formed mineralized nodules in vitro, and formed bone in an in vivo transplantation assay. Increased values were also found in three adults with recent fractures. CONCLUSIONS Osteoblast-lineage cells circulate in physiologically significant numbers, correlate with markers of bone formation, and are markedly higher during pubertal growth; therefore, they may represent a previously unrecognized circulatory component to the process of bone formation.
Collapse
|
38
|
|
39
|
Shimko DA, Burks CA, Dee KC, Nauman EA. Comparison ofin VitroMineralization by Murine Embryonic and Adult Stem Cells Cultured in an Osteogenic Medium. ACTA ACUST UNITED AC 2004; 10:1386-98. [PMID: 15588399 DOI: 10.1089/ten.2004.10.1386] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Nearly half a million bone-grafting procedures occurred in the United States in the year 2000. Tissue-engineered bone substitutes may mitigate difficulties associated with current grafting options. Embryonic stem cells (ESCs) could be a potential cell source for bone substitutes; however, direct comparisons between ESCs and other cell sources are lacking. Here we provide a direct, long-term, in vitro comparison of mineralization processes in adult, marrow-derived, mesenchymal stem cells (MSCs) and ESCs from the 129/Sv+c/+p mouse strain. MSCs were observed to grow at a slower rate than ESCs. MSCs expressed seven times more alkaline phosphatase (AP) per cell than did ESCs and immediately showed type I collagen and osteocalcin production. ESCs also produced type I collagen and osteocalcin, but production was delayed. Mineral deposition by ESCs was nearly 50 times higher than by MSCs. Spectroscopic analysis showed the calcium-to-phosphorus ratio (Ca:P) of the ESC mineral (1.26:1) to be significantly higher than that of the MSCs (0.29:1), but still 25% lower than hydroxyapatite (1.67:1). Addition of basic fibroblast growth factor significantly inhibited AP expression, mineral deposition, and Ca:P ratios in MSCs and had little effect on ESCs. These functional characteristics may assist with cell selection for purposes of bone tissue engineering.
Collapse
Affiliation(s)
- Daniel A Shimko
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana, USA
| | | | | | | |
Collapse
|