1
|
Liu H, Xu K, He Y, Huang F. Mitochondria in Multi-Directional Differentiation of Dental-Derived Mesenchymal Stem Cells. Biomolecules 2023; 14:12. [PMID: 38275753 PMCID: PMC10813276 DOI: 10.3390/biom14010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/03/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
The pursuit of tissue regeneration has fueled decades of research in regenerative medicine. Among the numerous types of mesenchymal stem cells (MSCs), dental-derived mesenchymal stem cells (DMSCs) have recently emerged as a particularly promising candidate for tissue repair and regeneration. In recent years, evidence has highlighted the pivotal role of mitochondria in directing and orchestrating the differentiation processes of DMSCs. Beyond mitochondrial energy metabolism, the multifaceted functions of mitochondria are governed by the mitochondrial quality control (MQC) system, encompassing biogenesis, autophagy, and dynamics. Notably, mitochondrial energy metabolism not only governs the decision to differentiate but also exerts a substantial influence on the determination of differentiation directions. Furthermore, the MQC system exerts a nuanced impact on the differentiation of DMSCs by finely regulating the quality and mass of mitochondria. The review aims to provide a comprehensive overview of the regulatory mechanisms governing the multi-directional differentiation of DMSCs, mediated by both mitochondrial energy metabolism and the MQC system. We also focus on a new idea based on the analysis of data from many research groups never considered before, namely, DMSC-based regenerative medicine applications.
Collapse
Affiliation(s)
| | | | - Yifan He
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510000, China; (H.L.); (K.X.)
| | - Fang Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510000, China; (H.L.); (K.X.)
| |
Collapse
|
2
|
Ibrahim AA, Yoneis A, Elsakka A, Elwany S. Fat enhanced leukocyte-platelet-rich fibrin versus fascia lata in endoscopic reconstruction of CSF leaks. Eur Arch Otorhinolaryngol 2023; 280:4141-4147. [PMID: 37191915 PMCID: PMC10382364 DOI: 10.1007/s00405-023-08010-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 05/08/2023] [Indexed: 05/17/2023]
Abstract
PURPOSE The aim of this study was to use a new biological active fat enhanced leukocyte-platelet-rich fibrin membrane (L-PRF) for skull base defect reconstruction and compare its validity and reliability with the time-honored fascia lata. METHODS This prospective study was conducted on 48 patients with spontaneous CSF leaks who were divided into 2 matched groups by stratified randomization, 24 patients in each group. In group A we performed multilayer repair using fat enhanced L-PRF membrane. In group B we used fascia lata for the multilayer repair. In both groups we enforced the repair with mucosal grafts/flaps. RESULTS The two groups were statistically matched for age, sex, intracranial pressure, and site and size of the skull base defect. There was no statistically significant difference between the two groups regarding the outcome of the repair or recurrence of CSF leak during the first postoperative year. Meningitis occurred in one patient in group B and was successfully treated. Another patient in group B developed thigh hematoma which resolved spontaneously. CONCLUSION The fat enhanced L-PRF membrane is a valid reliable option in repair of CSF leaks. The membrane is autologous, readily available, easily prepared, and has the advange of including stromal fat, stromal vascular fraction (SVF), and leukocyte-platelet-rich fibrin (L-PRF). The present study showed that fat enhanced L-PRF membrane is stable, non-absorbable, not liable to shrink or become necrotic, and can establish good seal of the skull base defect and further enhance the healing process. The use of the membrane also has the advantage of avoiding thigh incision and possible hematoma formation.
Collapse
Affiliation(s)
- Ahmed Aly Ibrahim
- Department of Otolaryngology, Alexandria Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Ahmed Yoneis
- Department of Otolaryngology, Alexandria Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Ahmed Elsakka
- Egyptian Foundation for Metabolic Researches, Alexandria, Egypt
| | - Samy Elwany
- Department of Otolaryngology, Alexandria Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
3
|
Hohenwallner K, Troppmair N, Panzenboeck L, Kasper C, El Abiead Y, Koellensperger G, Lamp LM, Hartler J, Egger D, Rampler E. Decoding Distinct Ganglioside Patterns of Native and Differentiated Mesenchymal Stem Cells by a Novel Glycolipidomics Profiling Strategy. JACS AU 2022; 2:2466-2480. [PMID: 36465531 PMCID: PMC9709940 DOI: 10.1021/jacsau.2c00230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 10/01/2022] [Accepted: 10/03/2022] [Indexed: 06/17/2023]
Abstract
Gangliosides are an indispensable glycolipid class concentrated on cell surfaces with a critical role in stem cell differentiation. Nonetheless, owing to the lack of suitable methods for scalable analysis covering the full scope of ganglioside molecular diversity, their mechanistic properties in signaling and differentiation remain undiscovered to a large extent. This work introduces a sensitive and comprehensive ganglioside assay based on liquid chromatography, high-resolution mass spectrometry, and multistage fragmentation. Complemented by an open-source data evaluation workflow, we provide automated in-depth lipid species-level and molecular species-level annotation based on decision rule sets for all major ganglioside classes. Compared to conventional state-of-the-art methods, the presented ganglioside assay offers (1) increased sensitivity, (2) superior structural elucidation, and (3) the possibility to detect novel ganglioside species. A major reason for the highly improved sensitivity is the optimized spectral readout based on the unique capability of two parallelizable mass analyzers for multistage fragmentation. We demonstrated the high-throughput universal capability of our novel analytical strategy by identifying 254 ganglioside species. As a proof of concept, 137 unique gangliosides were annotated in native and differentiated human mesenchymal stem cells including 78 potential cell-state-specific markers and 38 previously unreported gangliosides. A general increase of the ganglioside numbers upon differentiation was observed as well as cell-state-specific clustering based on the ganglioside species patterns. The combination of the developed glycolipidomics assay with the extended automated annotation tool enables comprehensive in-depth ganglioside characterization as shown on biological samples of interest. Our results suggest ganglioside patterns as a promising quality control tool for stem cells and their differentiation products. Additionally, we believe that our analytical workflow paves the way for probing glycolipid-based biochemical processes shedding light on the enigmatic processes of gangliosides and glycolipids in general.
Collapse
Affiliation(s)
- Katharina Hohenwallner
- Department
of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna 1090, Austria
- Vienna
Doctoral School in Chemistry (DoSChem), University of Vienna, Vienna 1090, Austria
| | - Nina Troppmair
- Department
of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna 1090, Austria
- Vienna
Doctoral School in Chemistry (DoSChem), University of Vienna, Vienna 1090, Austria
| | - Lisa Panzenboeck
- Department
of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna 1090, Austria
- Vienna
Doctoral School in Chemistry (DoSChem), University of Vienna, Vienna 1090, Austria
| | - Cornelia Kasper
- Institute
of Cell and Tissue Culture Technologies, University of Natural Resources and Life Sciences, Vienna 1190, Austria
| | - Yasin El Abiead
- Department
of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna 1090, Austria
| | - Gunda Koellensperger
- Department
of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna 1090, Austria
| | - Leonida M. Lamp
- Institute
of Pharmaceutical Sciences, University of
Graz, Graz 8010, Austria
| | - Jürgen Hartler
- Institute
of Pharmaceutical Sciences, University of
Graz, Graz 8010, Austria
- Field
of Excellence BioHealth − University
of Graz, Graz 8010, Austria
| | - Dominik Egger
- Institute
of Cell and Tissue Culture Technologies, University of Natural Resources and Life Sciences, Vienna 1190, Austria
| | - Evelyn Rampler
- Department
of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna 1090, Austria
| |
Collapse
|
4
|
Egger D, Lavrentieva A, Kugelmeier P, Kasper C. Physiologic isolation and expansion of human mesenchymal stem/stromal cells for manufacturing of cell-based therapy products. Eng Life Sci 2022; 22:361-372. [PMID: 35382547 PMCID: PMC8961040 DOI: 10.1002/elsc.202100097] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/13/2021] [Accepted: 10/15/2021] [Indexed: 01/04/2023] Open
Abstract
The utilization of mesenchymal stem/stromal cells raises new hopes in treatment of diseases and pathological conditions, while at the same time bringing immense challenges for researchers, manufacturers and physicians. It is essential to consider all steps along the in vitro fabrication of cell-based products in order to reach efficient and reproducible treatment outcomes. Here, the optimal protocols for isolation, cultivation and differentiation of mesenchymal stem cells are required. In this review we discuss these aspects and their influence on the final cell-based product quality. We demonstrate that physiological in vitro cell cultivation conditions play a crucial role in therapeutic functionalities of cultivated cells. We show that three-dimensional cell culture, dynamic culture conditions and physiologically relevant in vitro oxygen concentrations during isolation and expansion make a decisive contribution towards the improvement of cell-based products in regenerative medicine.
Collapse
Affiliation(s)
- Dominik Egger
- Department of BiotechnologyUniversity of Natural Resources and Life ScienceViennaAustria
| | | | | | - Cornelia Kasper
- Department of BiotechnologyUniversity of Natural Resources and Life ScienceViennaAustria
| |
Collapse
|
5
|
Effect of biomolecules derived from human platelet-rich plasma on the ex vivo expansion of human adipose-derived mesenchymal stem cells for clinical applications. Biologicals 2021; 75:37-48. [PMID: 34785135 DOI: 10.1016/j.biologicals.2021.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/02/2021] [Accepted: 11/06/2021] [Indexed: 11/20/2022] Open
Abstract
Mesenchymal stem cells are a tool in cell therapies but demand a large cell number per treatment, for that, suitable culture media is required which contains fetal bovine serum (FBS). However, for cell-based therapy applications, the use of FBS is problematic. Several alternatives to FBS have been explored, including human derivatives from platelet-rich plasma (hD-PRP). Although various studies have evaluated the impact of hD-PRP on MSC proliferation and differentiation, few of them have assessed their influence on processes, such as metabolism and gene expression. Here, we cultured human adipose-derived MSCs (hAD-MSCs) in media supplemented with either 10% hD-PRP (hD-PRP-SM) or 10% FBS (FBS-SM) in order to characterize them and evaluate the effect of hD-PRP on cell metabolism, gene expression of associated regenerative factors, as well as chromosome stability during cell expansion. We found that hAD-MSCs cultured in hD-PRP-SM have a greater cell elongation but express similar surface markers; in addition, hD-PRP-SM promoted a significant osteogenic differentiation in the absence of differentiation medium and increased the growth rate, maintaining chromosomal stability. In terms of cell metabolic profile, hAD-MSC behavior did not reveal any differences between both culture conditions. Conversely, significant differences in collagen I and angiopoietin 2 expression were observed between both conditions. The present results suggest that hD-PRP may influence hAD-MSC behavior.
Collapse
|
6
|
Born G, Nikolova M, Scherberich A, Treutlein B, García-García A, Martin I. Engineering of fully humanized and vascularized 3D bone marrow niches sustaining undifferentiated human cord blood hematopoietic stem and progenitor cells. J Tissue Eng 2021; 12:20417314211044855. [PMID: 34616539 PMCID: PMC8488506 DOI: 10.1177/20417314211044855] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/21/2021] [Indexed: 01/01/2023] Open
Abstract
Hematopoietic stem and progenitor cells (HSPCs) are frequently located around the bone marrow (BM) vasculature. These so-called perivascular niches regulate HSC function both in health and disease, but they have been poorly studied in humans due to the scarcity of models integrating complete human vascular structures. Herein, we propose the stromal vascular fraction (SVF) derived from human adipose tissue as a cell source to vascularize 3D osteoblastic BM niches engineered in perfusion bioreactors. We show that SVF cells form self-assembled capillary structures, composed by endothelial and perivascular cells, that add to the osteogenic matrix secreted by BM mesenchymal stromal cells in these engineered niches. In comparison to avascular osteoblastic niches, vascularized BM niches better maintain immunophenotypically-defined cord blood (CB) HSCs without affecting cell proliferation. In contrast, HSPCs cultured in vascularized BM niches showed increased CFU-granulocyte-erythrocyte-monocyte-megakaryocyte (CFU-GEMM) numbers. The vascularization also contributed to better preserve osteogenic gene expression in the niche, demonstrating that niche vascularization has an influence on both hematopoietic and stromal compartments. In summary, we have engineered a fully humanized and vascularized 3D BM tissue to model native human endosteal perivascular niches and revealed functional implications of this vascularization in sustaining undifferentiated CB HSPCs. This system provides a unique modular platform to explore hemato-vascular interactions in human healthy/pathological hematopoiesis.
Collapse
Affiliation(s)
- Gordian Born
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.,Department of Biomedical Engineering, University of Basel, Allschwill, Switzerland
| | - Marina Nikolova
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Arnaud Scherberich
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.,Department of Biomedical Engineering, University of Basel, Allschwill, Switzerland
| | - Barbara Treutlein
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Andrés García-García
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.,Department of Biomedical Engineering, University of Basel, Allschwill, Switzerland
| |
Collapse
|
7
|
Liu T, Xu J, Pan X, Ding Z, Xie H, Wang X, Xie H. Advances of adipose-derived mesenchymal stem cells-based biomaterial scaffolds for oral and maxillofacial tissue engineering. Bioact Mater 2021; 6:2467-2478. [PMID: 33553828 PMCID: PMC7850942 DOI: 10.1016/j.bioactmat.2021.01.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/03/2021] [Accepted: 01/11/2021] [Indexed: 02/05/2023] Open
Abstract
The management of oral and maxillofacial tissue defects caused by tumors, trauma, and congenital or acquired deformities has been a major challenge for surgeons over the last few decades. Autologous tissue transplantation, the gold standard of tissue reconstruction, is a valid method for repairing the oral and maxillofacial functions and aesthetics. However, several limitations hinder its clinical applications including complications of donor sites, limited tissue volume, and uncertain long-term outcomes. Adipose-derived mesenchymal stem cells (ADMSCs) widely exist in adipose tissue and can be easily obtained through liposuction. Like the bone marrow-derived mesenchymal stem cells (BMSCs), ADMSCs also have the multi-pluripotent potencies to differentiate into osteoblasts, chondrocytes, neurons, and myocytes. Therefore, the multilineage capacity of ADMSCs makes them valuable for cell-based medical therapies. In recent years, researchers have developed many candidates of ADMSCs-based biomaterial scaffolds to cater for the needs of oral and maxillofacial tissue engineering due to their superior performance. This review presents the advances and applications of ADMSCs-based biomaterial scaffolds, and explores their tissue engineering prospects in oral and maxillofacial reconstructions.
Collapse
Affiliation(s)
- Tong Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jia Xu
- The Key Laboratory of Oral Biomedicine, Jiangxi Province, School of Stomatology, Nanchang University, Nanchang, 330006, China
| | - Xun Pan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Zhangfan Ding
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Hao Xie
- General Surgery Department, The Second Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, 241000, China
| | - Xiaoyi Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Huixu Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
8
|
Filho DM, de Carvalho Ribeiro P, Oliveira LF, Dos Santos ALRT, Parreira RC, Pinto MCX, Resende RR. Enhancing the Therapeutic Potential of Mesenchymal Stem Cells with the CRISPR-Cas System. Stem Cell Rev Rep 2020; 15:463-473. [PMID: 31147819 DOI: 10.1007/s12015-019-09897-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Mesenchymal stem cells (MSCs), also known as multipotent mesenchymal stromal stem cells, are found in the perivascular space of several tissues. These cells have been subject of intense research in the last decade due to their low teratogenicity, as well as their ability to differentiate into mature cells and to secrete immunomodulatory and trophic factors. However, they usually promote only a modest benefit when transplanted in experimental disease models, one of the limitations for their clinical application. The CRISPR-Cas system, in turn, is highlighted as a simple and effective tool for genetic engineering. This system was tested in clinical trials over a relatively short period of time after establishing its applicability to the edition of the mammalian cell genome. Similar to the research evolution in MSCs, the CRISPR-Cas system demonstrated inconsistencies that limited its clinical application. In this review, we outline the evolution of MSC research and its applicability, and the progress of the CRISPR-Cas system from its discovery to the most recent clinical trials. We also propose perspectives on how the CRISPR-Cas system may improve the therapeutic potential of MSCs, making it more beneficial and long lasting.
Collapse
Affiliation(s)
- Daniel Mendes Filho
- Department of Physiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Patrícia de Carvalho Ribeiro
- Laboratory of Immunology and Experimental Transplantation, São José do Rio Preto Medical School, São José do Rio Preto, São Paulo, Brazil.,Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Lucas Felipe Oliveira
- Department of Physiology, Biological and Natural Sciences Institute, Triangulo Mineiro Federal University, Uberaba, Minas Gerais, Brazil.,National Institute of Science and Technology for Regenerative Medicine (INCT-REGENERA-CNPq), Rio de Janeiro, RJ, Brazil.,Minas Gerais Network for Tissue Engineering and Cell Therapy (REMETTECFAPEMIG), Belo Horizonte, MG, Brazil
| | | | - Ricardo Cambraia Parreira
- Department of Pharmacology, Biological Sciences Institute, Goias Federal University, Goiania, Goias, Brazil.
| | - Mauro Cunha Xavier Pinto
- Department of Pharmacology, Biological Sciences Institute, Goias Federal University, Goiania, Goias, Brazil
| | - Rodrigo Ribeiro Resende
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
9
|
Wang Y, Wang S, Gu C, Xiong Y, Shen H, Liu F, Yang J. Ex-vivo treatment of allografts using adipose-derived stem cells induced prolonged rejection-free survival in an allogenic hind-limb transplantation model. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:867. [PMID: 32793711 DOI: 10.21037/atm-19-4730] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Vascularized composite tissue allotransplantation (VCA) has increasingly been adopted for the reconstruction of tissues following severe injury. However, the side effects of the post-operative use of immunosuppressants may outweigh the benefits of VCA. In order to overcome this obstacle, ex-vivo pretreatment of allografts combined with mesenchymal stem cell-based therapy may help induce immunotolerance in composite tissue allotransplantation. Methods A hind-limb allotransplantation model of Brown-Norway to Lewis rats was established, and the allografts were infused with adipose-derived stem cells (ADSCs) and hypoxia primed ADSCs, which were injected through the vascular system along with short-term immunosuppressant treatment. The rejection-free survival of the allografts was monitored, and the histopathological examination of allografts was performed. The peripheral T lymphocytes and cytokines were analyzed using flow cytometry and ELISA, while Tregs infiltration in allotissue was detected using immunohistochemical staining (IHC). Results This study found that the ex-vivo treatment of allografts using ADSCs prolonged the survival of the allografts, compared with the medium control, suppressed the proliferation and infiltration of T lymphocytes and improved the secretion of immunomodulatory cytokines, such as IL-10, as well as induced regulatory T cells (Tregs) expression in the allografts. Conclusions The ex-vivo pretreatment of allografts using ADSCs may function as an important adjunctive therapy for the induction of immunotolerance in VCA.
Collapse
Affiliation(s)
- Yinmin Wang
- Department of Plastic and Reconstructive Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.,Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Shoubao Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Chuan Gu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yao Xiong
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Hua Shen
- Department of Plastic and Reconstructive Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Fei Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Jun Yang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Ehlert M, Radtke A, Jędrzejewski T, Roszek K, Bartmański M, Piszczek P. In Vitro Studies on Nanoporous, Nanotubular and Nanosponge-Like Titania Coatings, with the Use of Adipose-Derived Stem Cells. MATERIALS 2020; 13:ma13071574. [PMID: 32235354 PMCID: PMC7177883 DOI: 10.3390/ma13071574] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/18/2020] [Accepted: 03/26/2020] [Indexed: 12/21/2022]
Abstract
In vitro biological research on a group of amorphous titania coatings of different nanoarchitectures (nanoporous, nanotubular, and nanosponge-like) produced on the surface of Ti6Al4V alloy samples have been carried out, aimed at assessing their ability to interact with adipose-derived mesenchymal stem cells (ADSCs) and affect their activity. The attention has been drawn to the influence of surface coating architecture and its physicochemical properties on the ADSCs proliferation. Moreover, in vitro co-cultures: (1) fibroblasts cell line L929/ADSCs and (2) osteoblasts cell line MG-63/ADSCs on nanoporous, nanotubular and nanosponge-like TiO2 coatings have been studied. This allowed for evaluating the impact of the surface properties, especially roughness and wettability, on the creation of the beneficial microenvironment for co-cultures and/or enhancing differentiation potential of stem cells. Obtained results showed that the nanoporous surface is favorable for ADSCs, has great biointegrative properties, and supports the growth of co-cultures with MG-63 osteoblasts and L929 fibroblasts. Additionally, the number of osteoblasts seeded and cultured with ADSCs on TNT5 surface raised after 72-h culture almost twice when compared with the unmodified scaffold and by 30% when compared with MG-63 cells growing alone. The alkaline phosphatase activity of MG-63 osteoblasts co-cultured with ADSCs increased, that indirectly confirmed our assumptions that TNT-modified scaffolds create the osteogenic niche and enhance osteogenic potential of ADSCs.
Collapse
Affiliation(s)
- Michalina Ehlert
- Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Gagarina 7, 87-100 Toruń, Poland;
- Nano-implant Ltd. Gagarina 5/102, 87-100 Toruń, Poland
| | - Aleksandra Radtke
- Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Gagarina 7, 87-100 Toruń, Poland;
- Nano-implant Ltd. Gagarina 5/102, 87-100 Toruń, Poland
- Correspondence: (A.R.); (P.P.); Tel.: +48-600321294 (A.R.); Tel.: +48-607883357 (P.P.)
| | - Tomasz Jędrzejewski
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Lwowska 1, 87-100 Toruń, Poland; (K.R.); (T.J.)
| | - Katarzyna Roszek
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Lwowska 1, 87-100 Toruń, Poland; (K.R.); (T.J.)
| | - Michał Bartmański
- Faculty of Mechanical Engineering, Gdańsk University of Technology, Gabriela Narutowicza 11/12, 80-233 Gdańsk, Poland;
| | - Piotr Piszczek
- Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Gagarina 7, 87-100 Toruń, Poland;
- Nano-implant Ltd. Gagarina 5/102, 87-100 Toruń, Poland
- Correspondence: (A.R.); (P.P.); Tel.: +48-600321294 (A.R.); Tel.: +48-607883357 (P.P.)
| |
Collapse
|
11
|
Ehlert M, Roszek K, Jędrzejewski T, Bartmański M, Radtke A. Titania Nanofiber Scaffolds with Enhanced Biointegration Activity-Preliminary In Vitro Studies. Int J Mol Sci 2019; 20:E5642. [PMID: 31718064 PMCID: PMC6888681 DOI: 10.3390/ijms20225642] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/07/2019] [Accepted: 11/09/2019] [Indexed: 12/11/2022] Open
Abstract
The increasing need for novel bone replacement materials has been driving numerous studies on modifying their surface to stimulate osteogenic cells expansion and to accelerate bone tissue regeneration. The goal of the presented study was to optimize the production of titania-based bioactive materials with high porosity and defined nanostructure, which supports the cell viability and growth. We have chosen to our experiments TiO2 nanofibers, produced by chemical oxidation of Ti6Al4V alloy. Fibrous nanocoatings were characterized structurally (X-ray diffraction (XRD)) and morphologically (scanning electron microscopy (SEM)). The wettability of the coatings and their mechanical properties were also evaluated. We have investigated the direct influence of the modified titanium alloy surfaces on the survival and proliferation of mesenchymal stem cells derived from adipose tissue (ADSCs). In parallel, proliferation of bone tissue cells-human osteoblasts MG-63 and connective tissue cells - mouse fibroblasts L929, as well as cell viability in co-cultures (osteoblasts/ADSCs and fibroblasts/ADSCs has been studied. The results of our experiments proved that among all tested nanofibrous coatings, the amorphous titania-based ones were the most optimal scaffolds for the integration and proliferation of ADSCs, fibroblasts, and osteoblasts. Thus, we postulated these scaffolds to have the osteopromotional potential. However, from the co-culture experiments it can be concluded that ADSCs have the ability to functionalize the initially unfavorable surface, and make it suitable for more specialized and demanding cells.
Collapse
Affiliation(s)
- Michalina Ehlert
- Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Gagarina 7, 87-100 Toruń, Poland;
- Nano-Implant Ltd., Gagarina 5/102, 87-100 Toruń, Poland
| | - Katarzyna Roszek
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Lwowska 1, 87-100 Toruń, Poland; (K.R.); (T.J.)
| | - Tomasz Jędrzejewski
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Lwowska 1, 87-100 Toruń, Poland; (K.R.); (T.J.)
| | - Michał Bartmański
- Faculty of Mechanical Engineering, Gdańsk University of Technology, Gabriela Narutowicza 11/12, 80-233 Gdańsk, Poland;
| | - Aleksandra Radtke
- Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Gagarina 7, 87-100 Toruń, Poland;
- Nano-Implant Ltd., Gagarina 5/102, 87-100 Toruń, Poland
| |
Collapse
|
12
|
Kim W, Kim G. Collagen/bioceramic-based composite bioink to fabricate a porous 3D hASCs-laden structure for bone tissue regeneration. Biofabrication 2019; 12:015007. [DOI: 10.1088/1758-5090/ab436d] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
13
|
Weiss-Bilka HE, Meagher MJ, Gargac JA, Niebur GL, Roeder RK, Wagner DR. Mineral deposition and vascular invasion of hydroxyapatite reinforced collagen scaffolds seeded with human adipose-derived stem cells. Biomater Res 2019; 23:15. [PMID: 31641529 PMCID: PMC6796373 DOI: 10.1186/s40824-019-0167-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 09/30/2019] [Indexed: 12/15/2022] Open
Abstract
Background Collagen-based scaffolds reinforced with hydroxyapatite (HA) are an attractive choice for bone tissue engineering because their composition mimics that of bone. We previously reported the development of compression-molded collagen-HA scaffolds that exhibited high porosity, interconnected pores, and mechanical properties that were well-suited for surgical handling and fixation. The objective of this study was to investigate these novel collagen-HA scaffolds in combination with human adipose-derived stem cells (hASCs) as a template for bone formation in a subcutaneous athymic mouse model. Methods Collagen-HA scaffolds and collagen-only scaffolds were fabricated as previously described, and a clinically approved bone void filler was used as a control for the material. Constructs were seeded with hASCs and were pre-treated with either control or osteogenic media. A cell-free group was also included. Scaffolds were implanted subcutaneously in the backs of athymic nude mice for 8 weeks. Mineral deposition was quantified via micro-computed tomography. Histological and immunofluorescence images of the explants were used to analyze their vascular invasion, remodeling and cellularity. Results Cell-free collagen-HA scaffolds and those that were pre-seeded with osteogenically differentiated hASCs supported mineral deposition and vascular invasion at comparable rates, while cell-seeded constructs treated with the control medium showed lower mineralization after implantation. HA-reinforcement allowed collagen constructs to maintain their shape, provided improved cell-tissue-scaffold integration, and resulted in a more organized tissue when pre-treated in an osteogenic medium. Scaffold type and pre-treatment also determined osteoclast activity and therefore potential remodeling of the constructs. Conclusions The results of this study cumulatively indicate that treatment medium and scaffold composition direct mineralization and angiogenic tissue formation in an ectopic model. The data suggest that it may be necessary to match the scaffold with a particular cell type and cell-specific pre-treatment to achieve optimal bone formation.
Collapse
Affiliation(s)
- Holly E Weiss-Bilka
- 1Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Matthew J Meagher
- 1Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Joshua A Gargac
- 2School of Engineering, University of Mount Union, Alliance, OH 44601 USA
| | - Glen L Niebur
- 1Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556 USA.,3Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Ryan K Roeder
- 1Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556 USA.,3Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Diane R Wagner
- 4Department of Mechanical and Energy Engineering, Indiana University-Purdue University Indianapolis, 723 W. Michigan Ave SL260, Indianapolis, IN 46202 USA
| |
Collapse
|
14
|
Kunisch E, Gunnella F, Wagner S, Dees F, Maenz S, Bossert J, Jandt KD, Kinne RW. The poly (l-lactid-co-glycolide; PLGA) fiber component of brushite-forming calcium phosphate cement induces the osteogenic differentiation of human adipose tissue-derived stem cells. ACTA ACUST UNITED AC 2019; 14:055012. [PMID: 31465298 DOI: 10.1088/1748-605x/ab3544] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A brushite-forming calcium phosphate cement (CPC) was mechanically stabilized by addition of poly (l-lactid-co-glycolide; PLGA) fibers (≤10% w/w). It proved highly biocompatible and its fiber component enhanced bone formation in a sheep lumbar vertebroplasty model. However, possible effects on the osteogenic differentiation of resident mesenchymal stem cells (MSCs) remained unexplored. The present study used a novel approach, simultaneously analyzing the influence of a solid CPC scaffold and its relatively low PLGA proportion (a mimicry of natural bone) on osteogenic, chondrogenic, and adipogenic differentiation, as well as the pluripotency of human adipose tissue-derived mesenchymal stem cells (hASCs). hASCs were cultured on CPC discs with/without PLGA fibers (5% and 10%) in the absence of osteogenic medium for 3, 7, and 14 d. Gene expression of osteogenic markers (Runx2, osterix, alkaline phosphatase, collagen I, osteonectin, osteopontin, osteocalcin), chondrogenic markers (collagen II, Sox9, aggrecan), adipogenic markers (PPARG, Leptin, and FABP4), and pluripotency markers (Nanog, Tert, Rex) was analyzed by RT-PCR. The ability of hASCs to synthesize alkaline phosphatase was also evaluated. Cell number and viability were determined by fluorescein diacetate/propidium iodide staining. Compared to pure CPC, cultivation of hASCs on fiber-reinforced CPC transiently induced the gene expression of Runx2 and osterix (day 3), and long-lastingly augmented the expression of alkaline phosphatase (and its enzyme activity), collagen I, and osteonectin (until day 14). In contrast, augmented expression of all chondrogenic, adipogenic, and pluripotency markers was limited to day 3, followed by significant downregulation. Cultivation of hASCs on fiber-reinforced CPC reduced the cell number, but not the proportion of viable cells (viability > 95%). The PLGA component of fiber-reinforced, brushite-forming CPC supports long-lasting osteogenic differentiation of hASCs, whereas chondrogenesis, adipogenesis, and pluripotency are initially augmented, but subsequently suppressed. In view of parallel animal results, PLGA fibers may represent an interesting clinical target for future improvement of CPC- based bone regeneration.
Collapse
Affiliation(s)
- Elke Kunisch
- Experimental Rheumatology Unit, Department of Orthopedics, Jena University Hospital, Waldkliniken Eisenberg GmbH, Eisenberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
15
|
From 3D to 3D: isolation of mesenchymal stem/stromal cells into a three-dimensional human platelet lysate matrix. Stem Cell Res Ther 2019; 10:248. [PMID: 31399129 PMCID: PMC6688329 DOI: 10.1186/s13287-019-1346-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/02/2019] [Accepted: 07/15/2019] [Indexed: 12/19/2022] Open
Abstract
Background Mesenchymal stem/stromal cells (MSCs) are considered an important candidate in cell therapy and tissue engineering approaches. The culture of stem cells in a 3D environment is known to better resemble the in vivo situation and to promote therapeutically relevant effects in isolated cells. Therefore, the aim of this study was to develop an approach for the direct isolation of MSCs from adipose tissue into a 3D environment, avoiding contact to a 2D plastic surface. Furthermore, the use of a cryoprotective medium for the cryopreservation of whole adipose tissue was evaluated. Materials and methods Cryopreservation of fresh adipose tissue with and without a cryoprotective medium was compared with regard to the viability and metabolic activity of cells. After thawing, the tissue was embedded in a novel human platelet lysate-based hydrogel for the isolation of MSCs. The migration, yield, viability, and metabolic activity of cells from the 3D matrix were compared to cells from 2D explant culture. Also, the surface marker profile and differentiation capacity of MSCs from the 3D matrix were evaluated and compared to MSCs from isolation by enzymatic treatment or 2D explant culture. Results The cryopreservation of whole adipose tissue was found to be feasible, and therefore, adipose tissue can be stored and is available for MSC isolation on demand. Also, we demonstrate the isolation of MSCs from adipose tissue into the 3D matrix. The cells derived from this isolation procedure display a similar phenotype and differentiation capacity like MSCs derived by traditional procedures. Conclusions The presented approach allows to cryopreserve adipose tissue. Furthermore, for the first time, MSCs were directly isolated from the tissue into a soft 3D hydrogel environment, avoiding any contact to a 2D plastic culture surface. Electronic supplementary material The online version of this article (10.1186/s13287-019-1346-2) contains supplementary material, which is available to authorized users.
Collapse
|
16
|
Silva KR, Baptista LS. Adipose-derived stromal/stem cells from different adipose depots in obesity development. World J Stem Cells 2019; 11:147-166. [PMID: 30949294 PMCID: PMC6441940 DOI: 10.4252/wjsc.v11.i3.147] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 01/27/2019] [Accepted: 02/28/2019] [Indexed: 02/06/2023] Open
Abstract
The increasing prevalence of obesity is alarming because it is a risk factor for cardiovascular and metabolic diseases (such as type 2 diabetes). The occurrence of these comorbidities in obese patients can arise from white adipose tissue (WAT) dysfunctions, which affect metabolism, insulin sensitivity and promote local and systemic inflammation. In mammals, WAT depots at different anatomical locations (subcutaneous, preperitoneal and visceral) are highly heterogeneous in their morpho-phenotypic profiles and contribute differently to homeostasis and obesity development, depending on their ability to trigger and modulate WAT inflammation. This heterogeneity is likely due to the differential behavior of cells from each depot. Numerous studies suggest that adipose-derived stem/stromal cells (ASC; referred to as adipose progenitor cells, in vivo) with depot-specific gene expression profiles and adipogenic and immunomodulatory potentials are keys for the establishment of the morpho-functional heterogeneity between WAT depots, as well as for the development of depot-specific responses to metabolic challenges. In this review, we discuss depot-specific ASC properties and how they can contribute to the pathophysiology of obesity and metabolic disorders, to provide guidance for researchers and clinicians in the development of ASC-based therapeutic approaches.
Collapse
Affiliation(s)
- Karina Ribeiro Silva
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology, Duque de Caxias, RJ 25250-020, Brazil
- Post-Graduation Program of Biotechnology, National Institute of Metrology, Quality and Technology, Duque de Caxias, RJ 25250-020, Brazil
| | - Leandra Santos Baptista
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology, Duque de Caxias, RJ 25250-020, Brazil
- Post-Graduation Program of Biotechnology, National Institute of Metrology, Quality and Technology, Duque de Caxias, RJ 25250-020, Brazil
- Multidisciplinary Center for Biological Research (Numpex-Bio), Federal University of Rio de Janeiro Campus Duque de Caxias, Duque de Caxias, RJ 25245-390, Brazil
| |
Collapse
|
17
|
Ishihara M, Kishimoto S, Nakamura S, Fukuda K, Sato Y, Hattori H. Biomaterials as cell carriers for augmentation of adipose tissue-derived stromal cell transplantation. Biomed Mater Eng 2019; 29:567-585. [PMID: 30400072 DOI: 10.3233/bme-181009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Adipose tissue-derived stromal cells (ADSCs) contain lineage-committed progenitor cells that have the ability to differentiate into various cell types that may be useful for autologous cell transplantation to correct defects of skin, adipose, cartilage, bone, tendon, and blood vessels. The multipotent characteristics of ADSCs, as well as their abundance in the human body, make them an attractive potential resource for wound repair and applications to tissue engineering. ADSC transplantation has been used in combination with biomaterials, including cell sheets, hydrogel, and three-dimensional (3D) scaffolds based on chitosan, fibrin, atelocollagen, and decellularized porcine dermis, etc. Furthermore, low molecular weight heparin/protamine nanoparticles (LH/P NPs) have been used as an inducer of ADSC aggregation. The tissue engineering potential of these biomaterials as cell carriers is increased by the synergistic relationship between ADSCs and the biomaterials, resulting in the release of angiogenic cytokines and growth factors. In this review article, we describe the advantages of ADSC transplantation for tissue engineering, focusing on biomaterials as cell carriers which we have studied.
Collapse
Affiliation(s)
- Masayuki Ishihara
- Division of Biomedical Engineering Research Institute, National Defense Medical College, Saitama 359-8513, Japan
| | - Satoko Kishimoto
- Research Support Center, Dokkyo Medical University, Tochigi 321-0293, Japan
| | - Shingo Nakamura
- Division of Biomedical Engineering Research Institute, National Defense Medical College, Saitama 359-8513, Japan
| | - Koichi Fukuda
- Division of Biomedical Engineering Research Institute, National Defense Medical College, Saitama 359-8513, Japan
| | - Yoko Sato
- Division of Biomedical Engineering Research Institute, National Defense Medical College, Saitama 359-8513, Japan
| | - Hidemi Hattori
- Department of Biochemistry and Applied Sciences, University of Miyazaki, Miyazaki 889-2162, Japan
| |
Collapse
|
18
|
Dziedzic DSM, Mogharbel BF, Ferreira PE, Irioda AC, de Carvalho KAT. Transplantation of Adipose-derived Cells for Periodontal Regeneration: A Systematic Review. Curr Stem Cell Res Ther 2019; 14:504-518. [PMID: 30394216 DOI: 10.2174/1574888x13666181105144430] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/22/2018] [Accepted: 10/29/2018] [Indexed: 12/22/2022]
Abstract
This systematic review evaluated the transplantation of cells derived from adipose tissue for applications in dentistry. SCOPUS, PUBMED and LILACS databases were searched for in vitro studies and pre-clinical animal model studies using the keywords "ADIPOSE", "CELLS", and "PERIODONTAL", with the Boolean operator "AND". A total of 160 titles and abstracts were identified, and 29 publications met the inclusion criteria, 14 in vitro and 15 in vivo studies. In vitro studies demonstrated that adipose- derived cells stimulate neovascularization, have osteogenic and odontogenic potential; besides adhesion, proliferation and differentiation on probable cell carriers. Preclinical studies described improvement of bone and periodontal healing with the association of adipose-derived cells and the carrier materials tested: Platelet Rich Plasma, Fibrin, Collagen and Synthetic polymer. There is evidence from the current in vitro and in vivo data indicating that adipose-derived cells may contribute to bone and periodontal regeneration. The small quantity of studies and the large variation on study designs, from animal models, cell sources and defect morphology, did not favor a meta-analysis. Additional studies need to be conducted to investigate the regeneration variability and the mechanisms of cell participation in the processes. An overview of animal models, cell sources, and scaffolds, as well as new perspectives are provided for future bone and periodontal regeneration study designs.
Collapse
Affiliation(s)
- Dilcele Silva Moreira Dziedzic
- Pele Pequeno Principe Institute for Child and Adolescent Health Research, Pequeno Principe Faculty, Curitiba, Brazil
- Dentistry Faculty, Universidade Positivo, Curitiba, Brazil
| | - Bassam Felipe Mogharbel
- Pele Pequeno Principe Institute for Child and Adolescent Health Research, Pequeno Principe Faculty, Curitiba, Brazil
| | - Priscila Elias Ferreira
- Pele Pequeno Principe Institute for Child and Adolescent Health Research, Pequeno Principe Faculty, Curitiba, Brazil
| | - Ana Carolina Irioda
- Pele Pequeno Principe Institute for Child and Adolescent Health Research, Pequeno Principe Faculty, Curitiba, Brazil
| | | |
Collapse
|
19
|
MicroRNA 210 Mediates VEGF Upregulation in Human Periodontal Ligament Stem Cells Cultured on 3DHydroxyapatite Ceramic Scaffold. Int J Mol Sci 2018; 19:ijms19123916. [PMID: 30563289 PMCID: PMC6320762 DOI: 10.3390/ijms19123916] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 12/03/2018] [Accepted: 12/05/2018] [Indexed: 12/19/2022] Open
Abstract
The aim of the present research was the evaluation of the behavior of human periodontal ligament stem cells (hPDLSCs), cultured in presence of Endobon® Xenograft Granules (G), a fully deproteinated hydroxyapatite ceramic scaffold derived from cancellous bovine bone. hPDLSCs were seeded with and without G for 24 h to 1 week. The cell growth, morphological features, adhesiveness, differentiation ability, modulation of miR-210 and Vascular Endothelial Growth Factor (VEGF) secretion were analyzed by means of MTT assay, Scanning Electron Microscopy (SEM), Confocal Laser Scanning Microscopy (CLSM), Alizarin Red S assay, RT-PCR and ELISA test, respectively. hPDLSCs grown on the biomaterial showed the ability to form focal adhesion on the substrate, as demonstrated by vinculin expression. These data were supported by SEM analysis showing that an adhesiveness process associated to cell growth occurs between cells and biomaterials. The osteogenic differentiation, evaluated by morphological, biochemical, and RT-PCR analysis, was pronounced in the hPDLSCs grown in the three-dimensional inorganic bovine bone substitute in the presence of osteoinductive conditions. In addition, an upregulation of miR-210 and VEGF was evident in cells cultured in presence of the biomaterial. Our results inspire us to consider granules not only an adequate biocompatible three-dimensional biomaterial, but also an effective inductor of miR-210 and VEGF; in fact, the involvement of miR-210 in VEGF secretion could offer a novel regulatory system in the early steps of the bone-regeneration process.
Collapse
|
20
|
Du J, Xie P, Lin S, Wu Y, Zeng D, Li Y, Jiang X. Time-Phase Sequential Utilization of Adipose-Derived Mesenchymal Stem Cells on Mesoporous Bioactive Glass for Restoration of Critical Size Bone Defects. ACS APPLIED MATERIALS & INTERFACES 2018; 10:28340-28350. [PMID: 30080385 DOI: 10.1021/acsami.8b08563] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The effective transportation of oxygen, nutrients, and metabolic wastes through new blood vessel networks is key to the survival of engineered constructs in large bone defects. Adipose-derived mesenchymal stem cells (ADSCs), which are regarded as excellent candidates for both bone and blood vessel engineering, are the preferred option for the restoration of massive bone defects. Therefore, we propose to induce ADSCs into osteogenic and endothelial cells differently. A modified hierarchical mesoporous bioactive glass (MBG) scaffold with an enhanced compressive strength was constructed and prevascularized by seeding with endothelial-induced ADSCs (EI-ADSCs). The prevascularized scaffolds were combined with osteogenically induced ADSCs (OI-ADSCs) to repair critical-size bone defects. To validate the angiogenesis of the prevascularized MBG scaffolds in vivo, green fluorescent protein (GFP) was used to label EI-ADSCs. The labeled EI-ADSCs were demonstrated to survive and participate in vascularization at day 7 after subcutaneous implantation in nude mice by double immunofluorescence staining of CD31 and GFP. Regarding the restoration of critical size bone defects, early angiogenesis of rat femur plug defects was evaluated by perfusion of Microfil after 3 weeks. Compared to nonvascularized MBG carrying OI-ADSCs (MBG/OI-ADSCs) and non-cell-seeded MBG scaffolds, the prevascularized MBG carrying OI-ADSCs (Pv-MBG/OI-ADSCs) showed enhanced angiogenesis on the surface and interior. Through dynamic bone formation analysis with sequential fluorescent labeling and Van Gieson's picro-fuchsin staining, we found that the Pv-MBG/OI-ADSCs exhibited the highest mineral deposition rate after surgery, which may be contributed by rapid vascular anastomosis facilitating increased survival of the seeded OI-ADSCs and by the recruitment function for bone mesenchymal stem cells. Therefore, the strategy of time-phase sequential utilization of ADSCs on MBG scaffolds is a practical design for the repair of massive bone defects.
Collapse
Affiliation(s)
- Jiahui Du
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology , Shanghai Jiao Tong University School of Medicine , 639 Zhizaoju Road , Shanghai 200011 , China
- National Clinical Research Center for Oral Diseases , 639 Zhizaoju Road , Shanghai 200011 , China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology , 639 Zhizaoju Road , Shanghai 200011 , China
| | - Peng Xie
- The State Key Laboratory of Bioreactor Engineering, Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Center for Biomedical Materials of Ministry of Education , East China University of Science and Technology , Shanghai 200237 , China
| | - Shuxian Lin
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology , Shanghai Jiao Tong University School of Medicine , 639 Zhizaoju Road , Shanghai 200011 , China
- National Clinical Research Center for Oral Diseases , 639 Zhizaoju Road , Shanghai 200011 , China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology , 639 Zhizaoju Road , Shanghai 200011 , China
| | - Yuqiong Wu
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology , Shanghai Jiao Tong University School of Medicine , 639 Zhizaoju Road , Shanghai 200011 , China
- National Clinical Research Center for Oral Diseases , 639 Zhizaoju Road , Shanghai 200011 , China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology , 639 Zhizaoju Road , Shanghai 200011 , China
| | - Deliang Zeng
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology , Shanghai Jiao Tong University School of Medicine , 639 Zhizaoju Road , Shanghai 200011 , China
- National Clinical Research Center for Oral Diseases , 639 Zhizaoju Road , Shanghai 200011 , China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology , 639 Zhizaoju Road , Shanghai 200011 , China
| | - Yulin Li
- The State Key Laboratory of Bioreactor Engineering, Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Center for Biomedical Materials of Ministry of Education , East China University of Science and Technology , Shanghai 200237 , China
| | - Xinquan Jiang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology , Shanghai Jiao Tong University School of Medicine , 639 Zhizaoju Road , Shanghai 200011 , China
- National Clinical Research Center for Oral Diseases , 639 Zhizaoju Road , Shanghai 200011 , China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology , 639 Zhizaoju Road , Shanghai 200011 , China
| |
Collapse
|
21
|
Bothe F, Lotz B, Seebach E, Fischer J, Hesse E, Diederichs S, Richter W. Stimulation of calvarial bone healing with human bone marrow stromal cells versus inhibition with adipose-tissue stromal cells on nanostructured β-TCP-collagen. Acta Biomater 2018; 76:135-145. [PMID: 29933108 DOI: 10.1016/j.actbio.2018.06.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/11/2018] [Accepted: 06/15/2018] [Indexed: 12/28/2022]
Abstract
Bioactive functional scaffolds are essential for support of cell-based strategies to improve bone regeneration. Adipose-tissue-derived-stromal cells (ASC) are more accessible multipotent cells with faster proliferation than bone-marrow-derived-stromal-cells (BMSC) having potential to replace BMSC for therapeutic stimulation of bone-defect healing. Their osteogenic potential is, however, lower compared to BMSC, a deficit that may be overcome in growth factor-rich orthotopic bone defects with enhanced bone-conductive scaffolds. Objective of this study was to compare the therapeutic potency of human ASC and BMSC for bone regeneration on a novel nanoparticulate β-TCP/collagen-carrier (β-TNC). Cytotoxicity of β-TCP nanoparticles and multilineage differentiation of cells were characterized in vitro. Cell-seeded β-TNC versus cell-free controls were implanted into 4 mm calvarial bone-defects in immunodeficient mice and bone healing was quantified by µCT at 4 and 8 weeks. Tissue-quality and cell-origin were assessed by histology. β-TNC was non-toxic, radiolucent and biocompatible, lent excellent support for human cell persistence and allowed formation of human bone tissue by BMSC but not ASC. Opposite to BMSC, ASC-grafting significantly inhibited calvarial bone healing compared to controls. Bone formation progressed significantly from 4 to 8 weeks only in BMSC and controls yielding 5.6-fold more mineralized tissue in BMSC versus ASC-treated defects. Conclusively, β-TNC was simple to generate, biocompatible, osteoconductive, and stimulated osteogenicity of BMSC to enhance calvarial defect healing while ASC had negative effects. Thus, an orthotopic environment and β-TNC could not compensate for cell-autonomous deficits of ASC which should systematically be considered when choosing the right cell source for tissue engineering-based stimulation of bone regeneration. STATEMENT OF SIGNIFICANCE Bone-marrow-derived-stromal cells (BMSC) implanted on bone replacement materials can support bone defect healing and adipose-tissue-derived-stromal cells (ASC) being more accessible and better proliferating are considered as alternate source. This first standardized comparison of the bone regeneration potency of human ASC and BMSC was performed on a novel nanoparticular β-TCP-enriched collagen-carrier (β-TNC) designed to overcome the known inferior osteogenicity of ASC. β-TNC was non-toxic, biocompatible and osteoconductive supporting human bone formation and defect-closure by BMSC but not ASC. Long-term cell-persistence and the distinct secretome of ASC appear as main reasons why ASC inhibited bone healing opposite to BMSC. Overall, ASC-grafting is at considerable risk of producing negative effects on bone-healing while no such risks are known for BMSC.
Collapse
|
22
|
Maglione M, Salvador E, Ruaro ME, Melato M, Tromba G, Angerame D, Bevilacqua L. Bone regeneration with adipose derived stem cells in a rabbit model. J Biomed Res 2018; 33:38. [PMID: 30007953 PMCID: PMC6352878 DOI: 10.7555/jbr.32.20160066] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 03/07/2017] [Indexed: 01/22/2023] Open
Abstract
It has been shown that stem cells are able to calcify both in vitro and in vivo once implanted under the skin, if conveniently differentiated. Nowadays, however, a study on their efficiency in osseous regeneration does not exist in scientific literature and this very task is the real aim of the present experimentation. Five different defects of 6 mm in diameter and 2 mm in depth were created in the calvaria of 8 white New Zealand rabbits. Four defects were regenerated using 2 different conveniently modified scaffolds (Bio-Oss® Block and Bio-Oss Collagen®, Geistlich), with and without the aid of stem cells. After the insertion, the part was covered with a collagen membrane fixed by 5 modified titan pins (Altapin®). The defect in the front was left empty on purpose as an internal control to each animal. Two animals were sacrificed respectively after 2, 4, 6, 10 weeks. The samples were evaluated with micro-CT and histological analysis. Micro-CT analysis revealed that the quantity of new bone for samples with Bio-Oss® Block and stem cells was higher than for samples with Bio-Oss® Block alone. Histological analysis showed that regeneration occurred in an optimal way in every sample treated with scaffolds. The findings indicated that the use of adult stem cells combined with scaffolds accelerated some steps in normal osseous regeneration.
Collapse
Affiliation(s)
- Michele Maglione
- . Department of Medical Sciences, University of Trieste, Trieste 34125, Italy
| | - Enrico Salvador
- . Department of Medical Sciences, University of Trieste, Trieste 34125, Italy
| | - Maria E. Ruaro
- . SISSA-International School for Advanced Studies, Trieste 34136, Italy
| | - Mauro Melato
- . Department of Pathology and Legal Medicine, University of Trieste, Trieste 34125, Italy
| | - Giuliana Tromba
- . Elettra-Sincrotrone Trieste S.C.p.A., Trieste 34149, Italy
| | - Daniele Angerame
- . Department of Medical Sciences, University of Trieste, Trieste 34125, Italy
| | - Lorenzo Bevilacqua
- . Department of Medical Sciences, University of Trieste, Trieste 34125, Italy
| |
Collapse
|
23
|
Brennan MA, Renaud A, Guilloton F, Mebarki M, Trichet V, Sensebé L, Deschaseaux F, Chevallier N, Layrolle P. Inferior In Vivo Osteogenesis and Superior Angiogenesis of Human Adipose‐Derived Stem Cells Compared with Bone Marrow‐Derived Stem Cells Cultured in Xeno‐Free Conditions. Stem Cells Transl Med 2017; 6:2160-2172. [PMID: 29052365 PMCID: PMC5702520 DOI: 10.1002/sctm.17-0133] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/17/2017] [Indexed: 12/24/2022] Open
Abstract
The possibility of using adipose tissue-derived stromal cells (ATSC) as alternatives to bone marrow-derived stromal cells (BMSC) for bone repair has garnered interest due to the accessibility, high cell yield, and rapid in vitro expansion of ATSC. For clinical relevance, their bone forming potential in comparison to BMSC must be proven. Distinct differences between ATSC and BMSC have been observed in vitro and comparison of osteogenic potential in vivo is not clear to date. The aim of the current study was to compare the osteogenesis of human xenofree-expanded ATSC and BMSC in vitro and in an ectopic nude mouse model of bone formation. Human MSC were implanted with biphasic calcium phosphate biomaterials in subcutis pockets for 8 weeks. Implant groups were: BMSC, ATSC, BMSC and ATSC mixed together in different ratios, as well as MSC primed with either osteogenic supplements (250 μM ascorbic acid, 10 mM β-glycerolphosphate, and 10 nM dexamethasone) or 50 ng/ml recombinant bone morphogenetic protein 4 prior to implantation. In vitro results show osteogenic gene expression and differentiation potentials of ATSC. Despite this, ATSC failed to form ectopic bone in vivo, in stark contrast to BMSC, although osteogenic priming did impart minor osteogenesis to ATSC. Neovascularization was enhanced by ATSC compared with BMSC; however, less ATSC engrafted into the implant compared with BMSC. Therefore, in the content of bone regeneration, the advantages of ATSC over BMSC including enhanced angiogenesis, may be negated by their lack of osteogenesis and prerequisite for osteogenic differentiation prior to transplantation. Stem Cells Translational Medicine 2017;6:2160-2172.
Collapse
Affiliation(s)
- Meadhbh A. Brennan
- INSERM, UMR 1238, PHYOS, Laboratory of Bone Sarcomas and Remodelling of Calcified Tissues, Faculty of Medicine, University of NantesNantesFrance
| | - Audrey Renaud
- INSERM, UMR 1238, PHYOS, Laboratory of Bone Sarcomas and Remodelling of Calcified Tissues, Faculty of Medicine, University of NantesNantesFrance
| | - Fabien Guilloton
- STROMA Lab UMR UPS/CNRS 5273, U1031 INSERM, EFS‐Pyrénées‐MéditerranéeToulouseFrance
| | - Miryam Mebarki
- INSERM, IMRB U955‐E10, Engineering and Cellular Therapy Unit, Etablissement Français du Sang, Faculty of Medicine, Paris Est UniversityCréteilFrance
| | - Valerie Trichet
- INSERM, UMR 1238, PHYOS, Laboratory of Bone Sarcomas and Remodelling of Calcified Tissues, Faculty of Medicine, University of NantesNantesFrance
| | - Luc Sensebé
- STROMA Lab UMR UPS/CNRS 5273, U1031 INSERM, EFS‐Pyrénées‐MéditerranéeToulouseFrance
| | - Frederic Deschaseaux
- STROMA Lab UMR UPS/CNRS 5273, U1031 INSERM, EFS‐Pyrénées‐MéditerranéeToulouseFrance
| | - Nathalie Chevallier
- INSERM, IMRB U955‐E10, Engineering and Cellular Therapy Unit, Etablissement Français du Sang, Faculty of Medicine, Paris Est UniversityCréteilFrance
| | - Pierre Layrolle
- INSERM, UMR 1238, PHYOS, Laboratory of Bone Sarcomas and Remodelling of Calcified Tissues, Faculty of Medicine, University of NantesNantesFrance
| |
Collapse
|
24
|
Taghiyar L, Hosseini S, Hesaraki M, Azam Sayahpour F, Aghdami N, Baghaban Eslaminejad M. Isolation, Characterization and Osteogenic Potential of Mouse Digit Tip Blastema Cells in Comparison with Bone Marrow-Derived Mesenchymal Stem Cells In Vitro. CELL JOURNAL 2017; 19:585-598. [PMID: 29105393 PMCID: PMC5672097 DOI: 10.22074/cellj.2018.4710] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 11/02/2016] [Indexed: 12/20/2022]
Abstract
Objective Limb regeneration mediated by blastema cells (BlCs) in mammals is limited to the digit tips of neonates.
Due to the lack of access to BlCs in adults and the difficulty in isolating and expanding BlCs from neonates, the use
of a cellular population with similar features of BlCs would be a valuable strategy to direct a non-regenerative wound
towards regeneration. In this study, we have initially isolated and cultured BlCs, and explored their characteristics in
vitro. Next, we compared the capability of bone marrow-derived mesenchymal stem cells (BM-MSCs) as an alternative
accessible cell source to BlCs for regeneration of appendages.
Materials and Methods In this experimental study, BM-MSCs were isolated from BM and we obtained BlCs from the
neonatal regenerating digit tip of C57B/6 mice. The cells were characterized for expressions of cell surface markers by
flow cytometry. Quantitative-reverse transcription polymerase chain reaction (qRT-PCR) and lineage-specific staining
were used to assess their ability to differentiate into skeletal cell lineages. The colony forming ability, proliferation,
alkaline phosphatase (ALP) activity, calcium content, and osteogenic gene expression were evaluated in both BM-
MSCs and BlCs cultures at days 7, 14, and 21.
Results qRT-PCR analysis revealed that the cells from both sources readily differentiated into mesodermal lineages. There
was significantly higher colony forming ability in BM-MSCs compared to BlCs (P<0.05). Alizarin red staining (ARS), calcium,
and the ALP assay showed the same degree of mineral deposition in both BlCs and BM-MSCs. Gene expression levels of
osteblastic markers indicated similar bone differentiation capacity for both BlCs and BM-MSCs at all time-points.
Conclusion Characteristics of BlCs in vitro appear to be similar to BM-MSCs. Therefore, they could be considered as a
substitute for BlCs for a regenerative approach with potential use in future clinical settings for regenerating human appendages.
Collapse
Affiliation(s)
- Leila Taghiyar
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Samaneh Hosseini
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahdi Hesaraki
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Forough Azam Sayahpour
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nasser Aghdami
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
25
|
Yeo MG, Kim GH. A cell-printing approach for obtaining hASC-laden scaffolds by using a collagen/polyphenol bioink. Biofabrication 2017; 9:025004. [PMID: 28402968 DOI: 10.1088/1758-5090/aa6997] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
In the cell-printing process, bioink has been considered as an extremely important component for successful fabrication of macroscale cell-laden structures. Bioink should be non-toxic, biocompatible, and printable. To date, alginate has been widely used as a whole or partial component of bioink because it is non-toxic to embedded cells and even it can provide good printability with rapid gelation under calcium ions. However, alginate bioinks do not possess cell-activating ability. To overcome the shortcomings of alginate-based bioinks, a new collagen bioink, which was mixed with human adipose stem cells (hASCs) and crosslinked with a polyphenol (tannic acid), was proposed. The feasibility of the bioink was demonstrated using several in vitro assessments for comparison of the macroscale porous cell-laden collagen/polyphenol structure containing the hASCs with the conventional alginate-based cell-laden structure. The levels of the metabolic activity, including the cell viability and cell proliferation, of the cell-laden collagen structure were significantly higher than those of the control (alginate-based cell-laden structure). The results show that the newly designed bioink and cell-laden structure are potentially new outstanding components for regeneration of various tissues.
Collapse
Affiliation(s)
- Myung Gu Yeo
- Department of Biomechatronic Engineering, College of Biotechnology and Bioengineering, Sungkyunkwan University (SKKU), Suwon, Republic of Korea
| | | |
Collapse
|
26
|
Malec K, Góralska J, Hubalewska-Mazgaj M, Głowacz P, Jarosz M, Brzewski P, Sulka GD, Jaskuła M, Wybrańska I. Effects of nanoporous anodic titanium oxide on human adipose derived stem cells. Int J Nanomedicine 2016; 11:5349-5360. [PMID: 27789947 PMCID: PMC5072627 DOI: 10.2147/ijn.s116263] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The aim of current bone biomaterials research is to design implants that induce controlled, guided, successful, and rapid healing. Titanium implants are widely used in dental, orthopedic, and reconstructive surgery. A series of studies has indicated that cells can respond not only to the chemical properties of the biomaterial, but also, in particular, to the changes in surface topography. Nanoporous materials remain in focus of scientific queries due to their exclusive properties and broad applications. One such material is nanostructured titanium oxide with highly ordered, mutually perpendicular nanopores. Nanoporous anodic titanium dioxide (TiO2) films were fabricated by a three-step anodization process in propan-1,2,3-triol-based electrolyte containing fluoride ions. Adipose-derived stem cells offer many interesting opportunities for regenerative medicine. The important goal of tissue engineering is to direct stem cell differentiation into a desired cell lineage. The influence of nanoporous TiO2 with pore diameters of 80 and 108 nm on cell response, growth, viability, and ability to differentiate into osteoblastic lineage of human adipose-derived progenitors was explored. Cells were harvested from the subcutaneous abdominal fat tissue by a simple, minimally invasive, and inexpensive method. Our results indicate that anodic nanostructured TiO2 is a safe and nontoxic biomaterial. In vitro studies demonstrated that the nanotopography induced and enhanced osteodifferentiation of human adipose-derived stem cells from the abdominal subcutaneous fat tissue.
Collapse
Affiliation(s)
- Katarzyna Malec
- Department of Clinical Biochemistry, Jagiellonian University Medical College
| | - Joanna Góralska
- Department of Clinical Biochemistry, Jagiellonian University Medical College
| | - Magdalena Hubalewska-Mazgaj
- Department of Genetic Research and Nutrigenomics, Malopolska Centre of Biotechnology, Jagiellonian University
| | - Paulina Głowacz
- Department of Clinical Biochemistry, Jagiellonian University Medical College
| | - Magdalena Jarosz
- Department of Physical Chemistry and Electrochemistry, Faculty of Chemistry, Jagiellonian University
| | - Pawel Brzewski
- Department of Dermatology, Jagiellonian University Medical College, Kraków, Poland
| | - Grzegorz D Sulka
- Department of Physical Chemistry and Electrochemistry, Faculty of Chemistry, Jagiellonian University
| | - Marian Jaskuła
- Department of Physical Chemistry and Electrochemistry, Faculty of Chemistry, Jagiellonian University
| | - Iwona Wybrańska
- Department of Clinical Biochemistry, Jagiellonian University Medical College
| |
Collapse
|
27
|
Osinga R, Di Maggio N, Todorov A, Allafi N, Barbero A, Laurent F, Schaefer DJ, Martin I, Scherberich A. Generation of a Bone Organ by Human Adipose-Derived Stromal Cells Through Endochondral Ossification. Stem Cells Transl Med 2016; 5:1090-7. [PMID: 27334490 PMCID: PMC4954448 DOI: 10.5966/sctm.2015-0256] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 03/01/2016] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED : Recapitulation of endochondral ossification (ECO) (i.e., generation of marrow-containing ossicles through a cartilage intermediate) has relevance to develop human organotypic models for bone or hematopoietic cells and to engineer grafts for bone regeneration. Unlike bone marrow-derived stromal cells (also known as bone marrow-derived mesenchymal stromal/stem cells), adipose-derived stromal cells (ASC) have so far failed to form a bone organ by ECO. The goal of the present study was to assess whether priming human ASC to a defined stage of chondrogenesis in vitro allows their autonomous ECO upon ectopic implantation. ASC were cultured either as micromass pellets or into collagen sponges in chondrogenic medium containing transforming growth factor-β3 and bone morphogenetic protein-6 for 4 weeks (early hypertrophic templates) or for two additional weeks in medium supplemented with β-glycerophosphate, l-thyroxin, and interleukin1-β to induce hypertrophic maturation (late hypertrophic templates). Constructs were implanted in vivo and analyzed after 8 weeks. In vitro, ASC deposited cartilaginous matrix positive for glycosaminoglycans, type II collagen, and Indian hedgehog. Hypertrophic maturation induced upregulation of type X collagen, bone sialoprotein, and matrix metalloproteinase13 (MMP13). In vivo, both early and late hypertrophic templates underwent cartilage remodeling, as assessed by MMP13- and tartrate-resistant acid phosphatase-positive staining, and developed bone ossicles, including bone marrow elements, although to variable degrees of efficiency. In situ hybridization for human-specific sequences and staining with a human specific anti-CD146 antibody demonstrated the direct contribution of ASC to bone and stromal tissue formation. In conclusion, despite their debated skeletal progenitor nature, human ASC can generate bone organs through ECO when suitably primed in vitro. SIGNIFICANCE Recapitulation of endochondral ossification (ECO) (i.e., generation of marrow-containing ossicles through a cartilage intermediate) has relevance to develop human organotypic models for bone or hematopoietic cells and to engineer grafts for bone regeneration. This study demonstrated that expanded, human adult adipose-derived stromal cells can generate ectopic bone through ECO, as previously reported for bone marrow stromal cells. This system can be used as a model in a variety of settings for mimicking ECO during development, physiology, or pathology (e.g., to investigate the role of BMPs, their receptors, and signaling pathways). The findings have also translational relevance in the field of bone regeneration, which, despite several advances in the domains of materials and surgical techniques, still faces various limitations before being introduced in the routine clinical practice.
Collapse
Affiliation(s)
- Rik Osinga
- Department of Plastic, Reconstructive, Aesthetic, and Hand Surgery, University Hospital of Basel, Basel, Switzerland Laboratory of Tissue Engineering, Department of Surgery, University Hospital of Basel, Basel, Switzerland Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Nunzia Di Maggio
- Laboratory of Tissue Engineering, Department of Surgery, University Hospital of Basel, Basel, Switzerland Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Atanas Todorov
- Laboratory of Tissue Engineering, Department of Surgery, University Hospital of Basel, Basel, Switzerland Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Nima Allafi
- Department of Plastic, Reconstructive, Aesthetic, and Hand Surgery, University Hospital of Basel, Basel, Switzerland
| | - Andrea Barbero
- Laboratory of Tissue Engineering, Department of Surgery, University Hospital of Basel, Basel, Switzerland Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Frédéric Laurent
- Department of Biomedicine, University of Basel, Basel, Switzerland Developmental Genetics, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Dirk Johannes Schaefer
- Department of Plastic, Reconstructive, Aesthetic, and Hand Surgery, University Hospital of Basel, Basel, Switzerland
| | - Ivan Martin
- Laboratory of Tissue Engineering, Department of Surgery, University Hospital of Basel, Basel, Switzerland Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Arnaud Scherberich
- Laboratory of Tissue Engineering, Department of Surgery, University Hospital of Basel, Basel, Switzerland Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
28
|
Hwang IS, Bae HK, Cheong HT. Comparison of the characteristics and multipotential and in vivo cartilage formation capabilities between porcine adipose-derived stem cells and porcine skin-derived stem cell-like cells. Am J Vet Res 2016; 76:814-21. [PMID: 26309110 DOI: 10.2460/ajvr.76.9.814] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To compare the characteristics and multipotential and in vivo cartilage formation capabilities of porcine adipose-derived stem cells (pASCs) with those of porcine skin-derived stem cell-like cells (pSSCs). ANIMALS Three 6-month-old female pigs and four 6-week-old female athymic mice. PROCEDURES Adipose and skin tissue specimens were obtained from each pig following slaughter and digested to obtain pASCs and pSSCs. For each cell type, flow cytometry and reverse transcription PCR assays were performed to characterize the expression of cell surface and mesenchymal stem cell markers, and in vitro cell cultures were performed to determine the adipogenic, osteogenic, and chondrogenic capabilities. Each cell type was then implanted into athymic mice to determine the extent of in vivo cartilage formation after 6 weeks. RESULTS The cell surface and mesenchymal stem cell marker expression patterns, multipotential capability, and extent of in vivo cartilage formation did not differ significantly between pASCs and pSSCs. CONCLUSIONS AND CLINICAL RELEVANCE Results suggested that pSSCs may be a viable alternative to pASCs as a source of progenitor cells for tissue engineering in regenerative medicine.
Collapse
|
29
|
Quarto N, Senarath-Yapa K, Renda A, Longaker MT. TWIST1 silencing enhances in vitro and in vivo osteogenic differentiation of human adipose-derived stem cells by triggering activation of BMP-ERK/FGF signaling and TAZ upregulation. Stem Cells 2015; 33:833-47. [PMID: 25446627 DOI: 10.1002/stem.1907] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 10/06/2014] [Accepted: 10/15/2014] [Indexed: 01/10/2023]
Abstract
Mesenchymal stem cells (MSCs) show promise for cellular therapy and regenerative medicine. Human adipose tissue-derived stem cells (hASCs) represent an attractive source of seed cells in bone regeneration. How to effectively improve osteogenic differentiation of hASCs in the bone tissue engineering has become a very important question with profound translational implications. Numerous regulatory pathways dominate osteogenic differentiation of hASCs involving transcriptional factors and signaling molecules. However, how these factors combine with each other to regulate hASCs osteogenic differentiation still remains to be illustrated. The highly conserved developmental proteins TWIST play key roles for transcriptional regulation in mesenchymal cell lineages. This study investigates TWIST1 function in hASCs osteogenesis. Our results show that TWIST1 shRNA silencing increased the osteogenic potential of hASCs in vitro and their skeletal regenerative ability when applied in vivo. We demonstrate that the increased osteogenic capacity observed with TWIST1 knockdown in hASCs is mediated through endogenous activation of BMP and ERK/FGF signaling leading, in turn, to upregulation of TAZ, a transcriptional modulator of MSCs differentiation along the osteoblast lineage. Inhibition either of BMP or ERK/FGF signaling suppressed TAZ upregulation and the enhanced osteogenesis in shTWIST1 hASCs. Cosilencing of both TWIST1 and TAZ abrogated the effect elicited by TWIST1 knockdown thus, identifying TAZ as a downstream mediator through which TWIST1 knockdown enhanced osteogenic differentiation in hASCs. Our functional study contributes to a better knowledge of molecular mechanisms governing the osteogenic ability of hASCs, and highlights TWIST1 as a potential target to facilitate in vivo bone healing.
Collapse
Affiliation(s)
- Natalina Quarto
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, California, USA; Dipartimento di Scienze Biomediche Avanzate, Universita' degli Studi di Napoli Federico II, Napoli, Italy
| | | | | | | |
Collapse
|
30
|
Chuenjitkuntaworn B, Osathanon T, Nowwarote N, Supaphol P, Pavasant P. The efficacy of polycaprolactone/hydroxyapatite scaffold in combination with mesenchymal stem cells for bone tissue engineering. J Biomed Mater Res A 2015; 104:264-71. [DOI: 10.1002/jbm.a.35558] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 08/12/2015] [Accepted: 09/03/2015] [Indexed: 12/27/2022]
Affiliation(s)
| | - Thanaphum Osathanon
- Mineralized Tissue Research Unit, Faculty of Dentistry; Chulalongkorn University; Bangkok 10330 Thailand
- Department of Anatomy, Faculty of Dentistry; Chulalongkorn University; Bangkok Pathumwan 10330 Thailand
| | - Nunthawan Nowwarote
- Mineralized Tissue Research Unit, Faculty of Dentistry; Chulalongkorn University; Bangkok 10330 Thailand
| | - Pitt Supaphol
- The Petroleum and Petrochemical College; Chulalongkorn University; Bangkok Pathumwan 10330 Thailand
| | - Prasit Pavasant
- Mineralized Tissue Research Unit, Faculty of Dentistry; Chulalongkorn University; Bangkok 10330 Thailand
- Department of Anatomy, Faculty of Dentistry; Chulalongkorn University; Bangkok Pathumwan 10330 Thailand
| |
Collapse
|
31
|
Abuna RP, De Oliveira FS, Santos TDS, Guerra TR, Rosa AL, Beloti MM. Participation of TNF-α in Inhibitory Effects of Adipocytes on Osteoblast Differentiation. J Cell Physiol 2015; 231:204-14. [DOI: 10.1002/jcp.25073] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 06/05/2015] [Indexed: 01/16/2023]
Affiliation(s)
- Robrigo P.F. Abuna
- Cell Culture Laboratory; School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto; São Paulo Brazil
| | - Fabiola S. De Oliveira
- Cell Culture Laboratory; School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto; São Paulo Brazil
| | - Thiago De S. Santos
- Cell Culture Laboratory; School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto; São Paulo Brazil
| | - Thais R. Guerra
- Cell Culture Laboratory; School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto; São Paulo Brazil
| | - Adalberto L. Rosa
- Cell Culture Laboratory; School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto; São Paulo Brazil
| | - Marcio M. Beloti
- Cell Culture Laboratory; School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto; São Paulo Brazil
| |
Collapse
|
32
|
Bionaz M, Monaco E, Wheeler MB. Transcription Adaptation during In Vitro Adipogenesis and Osteogenesis of Porcine Mesenchymal Stem Cells: Dynamics of Pathways, Biological Processes, Up-Stream Regulators, and Gene Networks. PLoS One 2015; 10:e0137644. [PMID: 26398344 PMCID: PMC4580618 DOI: 10.1371/journal.pone.0137644] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 07/27/2015] [Indexed: 12/20/2022] Open
Abstract
The importance of mesenchymal stem cells (MSC) for bone regeneration is growing. Among MSC the bone marrow-derived stem cells (BMSC) are considered the gold standard in tissue engineering and regenerative medicine; however, the adipose-derived stem cells (ASC) have very similar properties and some advantages to be considered a good alternative to BMSC. The molecular mechanisms driving adipogenesis are relatively well-known but mechanisms driving osteogenesis are poorly known, particularly in pig. In the present study we have used transcriptome analysis to unravel pathways and biological functions driving in vitro adipogenesis and osteogenesis in BMSC and ASC. The analysis was performed using the novel Dynamic Impact Approach and functional enrichment analysis. In addition, a k-mean cluster analysis in association with enrichment analysis, networks reconstruction, and transcription factors overlapping analysis were performed in order to uncover the coordination of biological functions underlining differentiations. Analysis indicated a larger and more coordinated transcriptomic adaptation during adipogenesis compared to osteogenesis, with a larger induction of metabolism, particularly lipid synthesis (mostly triglycerides), and a larger use of amino acids for synthesis of feed-forward adipogenic compounds, larger cell signaling, lower cell-to-cell interactions, particularly for the cytoskeleton organization and cell junctions, and lower cell proliferation. The coordination of adipogenesis was mostly driven by Peroxisome Proliferator-activated Receptors together with other known adipogenic transcription factors. Only a few pathways and functions were more induced during osteogenesis compared to adipogenesis and some were more inhibited during osteogenesis, such as cholesterol and protein synthesis. Up-stream transcription factor analysis indicated activation of several lipid-related transcription regulators (e.g., PPARs and CEBPα) during adipogenesis but osteogenesis was driven by inhibition of several up-stream regulators, such as MYC. Between MSCs the data indicated an ‘adipocyte memory’ in ASC with also an apparent lower immunogenicity compared to BMSC during differentiations. Overall the analysis allowed proposing a dynamic model for the adipogenic and osteogenic differentiation in porcine ASC and BMSC.
Collapse
Affiliation(s)
- Massimo Bionaz
- Laboratory of Stem Cell Biology and Engineering in the Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Elisa Monaco
- Laboratory of Stem Cell Biology and Engineering in the Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Matthew B. Wheeler
- Laboratory of Stem Cell Biology and Engineering in the Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
33
|
Ishihara M, Kishimoto S, Takikawa M, Hattori H, Nakamura S, Shimizu M. Biomedical application of low molecular weight heparin/protamine nano/micro particles as cell- and growth factor-carriers and coating matrix. Int J Mol Sci 2015; 16:11785-803. [PMID: 26006248 PMCID: PMC4463730 DOI: 10.3390/ijms160511785] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 05/07/2015] [Accepted: 05/15/2015] [Indexed: 12/22/2022] Open
Abstract
Low molecular weight heparin (LMWH)/protamine (P) nano/micro particles (N/MPs) (LMWH/P N/MPs) were applied as carriers for heparin-binding growth factors (GFs) and for adhesive cells including adipose-derived stromal cells (ADSCs) and bone marrow-derived mesenchymal stem cells (BMSCs). A mixture of LMWH and P yields a dispersion of N/MPs (100 nm–3 μm in diameter). LMWH/P N/MPs can be immobilized onto cell surfaces or extracellular matrix, control the release, activate GFs and protect various GFs. Furthermore, LMWH/P N/MPs can also bind to adhesive cell surfaces, inducing cells and LMWH/P N/MPs-aggregate formation. Those aggregates substantially promoted cellular viability, and induced vascularization and fibrous tissue formation in vivo. The LMWH/P N/MPs, in combination with ADSCs or BMSCs, are effective cell-carriers and are potential promising novel therapeutic agents for inducing vascularization and fibrous tissue formation in ischemic disease by transplantation of the ADSCs and LMWH/P N/MPs-aggregates. LMWH/P N/MPs can also bind to tissue culture plates and adsorb exogenous GFs or GFs from those cells. The LMWH/P N/MPs-coated matrix in the presence of GFs may provide novel biomaterials that can control cellular activity such as growth and differentiation. Furthermore, three-dimensional (3D) cultures of cells including ADSCs and BMSCs using plasma-medium gel with LMWH/P N/MPs exhibited efficient cell proliferation. Thus, LMWH/P N/MPs are an adequate carrier both for GFs and for stromal cells such as ADSCs and BMSCs, and are a functional coating matrix for their cultures.
Collapse
Affiliation(s)
- Masayuki Ishihara
- Division of Biomedical Engineering Research Institute, National Defense Medical College, Saitama 359-8513, Japan.
| | - Satoko Kishimoto
- Research Support Center, Dokkyo Medical University, Tochigi 321-0293, Japan.
| | - Makoto Takikawa
- Department of Medical Engineering, National Defense Medical College, Saitama 359-8513, Japan.
| | - Hidemi Hattori
- Division of Biomedical Engineering Research Institute, National Defense Medical College, Saitama 359-8513, Japan.
| | - Shingo Nakamura
- Division of Biomedical Engineering Research Institute, National Defense Medical College, Saitama 359-8513, Japan.
| | - Masafumi Shimizu
- Department of Surgery, Tokorozawa Meisei Hospital, Saitama 359-1145, Japan.
| |
Collapse
|
34
|
Fanganiello RD, Ishiy FAA, Kobayashi GS, Alvizi L, Sunaga DY, Passos-Bueno MR. Increased In Vitro Osteopotential in SHED Associated with Higher IGF2 Expression When Compared with hASCs. Stem Cell Rev Rep 2015; 11:635-44. [DOI: 10.1007/s12015-015-9592-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
35
|
Hattori H, Ishihara M. Altered protein secretions during interactions between adipose tissue- or bone marrow-derived stromal cells and inflammatory cells. Stem Cell Res Ther 2015; 6:70. [PMID: 25884474 PMCID: PMC4417284 DOI: 10.1186/s13287-015-0052-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 01/04/2015] [Accepted: 03/13/2015] [Indexed: 12/21/2022] Open
Abstract
Introduction Paracrine effects can be exploited in cell-based therapies that secrete factors, such as chemokines and cytokines, and can recruit inflammatory cells to transplants. In this study, mouse adipose tissue-derived stromal cells (ASCs) and bone marrow-derived stromal cells (ST2 cells) were used to examine changes in paracrine interactions with inflammation cells. Methods Green fluorescent protein positive (GFP+) bone marrow cells (BMCs) were injected into an irradiated mouse via the femoral vein, and ASCs and ST2 cells were transplanted intradermally. Subsequently, an in vivo imaging system was used to observe behaviors of GFP+ BMCs. To detect bone marrow-derived inflammatory cells which migrated to the ASC and ST2 cell transplantation area, the sections were immunostained using antibodies against Gr1, CD11c, and F4/80, and secretory proteins were detected in culture medium using enzyme-linked immunosorbent assay. Results Many bone marrow-derived inflammatory cells migrated to ASC and ST2 cell transplantation sites. Among these, neutrophils were detected during the early period and macrophages were predominantly detected at a later point in time. Many chemokines, cytokines, growth factors, matrix metalloproteinases (MMPs), and tissue inhibitors of metalloproteinases (TIMPs) were secreted in abundance from ASCs, and the secretion increased by co-culturing with inflammatory cells, except for secretions of insulin-like growth factor-1, MMP-9 and MMP-13. Although secretions from ST2 cells were less than those from ASCs, co-culture with inflammatory cells increased these secretions to levels similar to those of ASCs. However, unlike ASCs, the ST2 cells did not secrete angiostatin, MMP-2, or MMP-3. Finally, ASCs secreted not only proinflammatory cytokines, angiogenic factors and MMPs but also anti-inflammatory cytokines, anti-angiogenesis factors, and TIMPs. Conclusions The effects of cell-based therapies using ASCs and ST2 cells are depended on paracrine effects that are mediated by chemokines, cytokines, growth factors, MMPs, and TIMPs, which comprise responses to interactions between transplanted cells and inflammatory cells. Moreover, paracrine effects of transplanted cells are influenced by inflammatory cells, and are moderated by a balance of secreted inhibitors. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0052-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hidemi Hattori
- Division of Biomedical Engineering, Research Institute, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan.
| | - Masayuki Ishihara
- Division of Biomedical Engineering, Research Institute, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan.
| |
Collapse
|
36
|
Adipose-Derived Stem Cells for Therapeutic Applications. Regen Med 2015. [DOI: 10.1007/978-1-4471-6542-2_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
37
|
Indumathi S, Mishra R, Harikrishnan R, Dhanasekaran M. Subcutaneous Adipose Tissue-Derived Stem Cells: Advancement and Applications in Regenerative Medicine. Regen Med 2015. [DOI: 10.1007/978-1-4471-6542-2_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
38
|
In Vitro Osteoinductive Effects of Hydroxycholesterol on Human Adipose-Derived Stem Cells Are Mediated through the Hedgehog Signaling Pathway. Plast Reconstr Surg 2014; 134:960-968. [DOI: 10.1097/prs.0000000000000601] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
39
|
Wang G, Zeng G, Wang C, Wang H, Yang B, Guan F, Li D, Feng X. Biocompatibility of quantum dots (CdSe/ZnS ) in human amniotic membrane-derived mesenchymal stem cells in vitro. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2014; 159:227-33. [PMID: 25277490 DOI: 10.5507/bp.2014.045] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 08/13/2014] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND AND AIM Amniotic membrane-derived mesenchymal stem cells (hAM-dMSCs) are a potential source of mesenchymal stem cells which could be used to repair skin damage. The use of mesenchymal stem cells to repair skin damage requires safe, effective and biocompatible agents to evaluate the effectiveness of the result. Quantum dots (QDs) composed of CdSe/ZnS are semiconductor nanocrystals with broad excitation and narrow emission spectra, which have been considered as a new chemical and fluorescent substance for non-invasively labeling different cells in vitro and in vivo. This study investigated the cytotoxic effects of QDs on hAM-dMSCs at different times following labeling. METHODS Using 0.75, 1.5 and 3.0 μL between quantum dots, labeled human amniotic mesenchymal stem cells were collected on days 1, 2 and 4 and observed morphological changes, performed an MTT cell growth assay and flow cytometry for mesenchymal stem cells molecular markers. RESULTS Quantum dot concentration 0.75 μg/mL labeled under a fluorescence microscope, cell morphology was observed, The MTT assay showed cells in the proliferative phase. Flow cytometry expression CD29, CD31, CD34, CD44, CD90, CD105 and CD106. CONCLUSIONS Within a certain range of concentrations between quantum dots labeled human amniotic mesenchymal stem cells has good biocompatibility.
Collapse
Affiliation(s)
| | | | - Caie Wang
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Science and Technology, LuoYang 471003 , Henan Province, PR China
| | - Huasheng Wang
- Department of Colorectal Surgery, People Hospital of Zhengzhou, Zhengzhou 450000 , Henan Province, PR China
| | - Bo Yang
- Department of Neurosurgery, the First Affiliated Hospital of ZhengZhou University, ZhengZho 450001, Henan Province, PR China
| | - Fangxia Guan
- Henan Academy of Medical Sciences, ZhengZhou 450001, Henan Province, PR China
| | - Dongpeng Li
- Department of Emergency, the First Affiliated Hospital of Henan University of Science and Technology, LuoYang 471003 , Henan Province, PR China
| | - Xiaoshan Feng
- Department of Oncological Surgery, the First Affiliated Hospital of Henan University of Science and Technology, LuoYang 471003 , Henan Province, PR China
| |
Collapse
|
40
|
In vivo differentiation of undifferentiated human adipose tissue-derived mesenchymal stem cells in critical-sized calvarial bone defects. Ann Plast Surg 2014; 72:225-33. [PMID: 23221992 DOI: 10.1097/sap.0b013e31825f70f5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Adult stem cells have recently drawn considerable attention for potential cell therapy applications. However, critical details about their specific in vivo environments and cellular activities are unclear. Adipose tissue-derived mesenchymal stem cells (ASCs) are attractive candidates for treating bone defects, but most studies focus on delivery of in vitro-differentiated cells. We assessed various scaffolding materials for the ability to support osteogenic differentiation of undifferentiated human ASCs in vivo, in athymic nude rat calvaria. Twenty-four 9- to 10-week-old athymic nude Sprague-Dawley rats (250 g) were used in these experiments. Fat tissue from 3 patients was harvested from abdominal tissue discarded during reconstructive breast surgery by transverse rectus abdominis myocutaneous flap, performed at the Asan Medical Center after resection of breast cancer. Human ASCs were extracted from discarded adipose tissue and isolated based on standard International Society for Cellular Therapy protocols. Adipose tissue-derived mesenchymal stem cells were seeded on polylactic glycolic acid, atelocollagen, and hydroxyapatite scaffolds, and osteogenesis was evaluated using bone mineral densitometry, histology, immunohistochemistry, and reverse transcription polymerase chain reaction. The gross appearance of scaffolds seeded with ASCs was strikingly different from that of scaffolds alone. Bone mineral densitometry analysis revealed a 2- to 3-fold increase in mineral density in ASC-seeded scaffolds. In addition, undifferentiated ASCs seeded onto hydroxyapatite scaffolds, but not onto collagen or polylactic glycolic acid scaffolds, expressed human messenger RNA for osteogenic markers such as alkaline phosphatase, osteopontin, osteocalcin, and osteonectin. These results indicate that undifferentiated human ASCs can differentiate into osteocytes or osteoblasts in athymic nude rat calvaria, and the importance of appropriate scaffolding for in vivo ASC differentiation.
Collapse
|
41
|
Effective wound healing in streptozotocin-induced diabetic rats by adipose-derived stromal cell transplantation in plasma-gel containing fragmin/protamine microparticles. Ann Plast Surg 2014; 72:113-20. [PMID: 24317245 DOI: 10.1097/sap.0000000000000014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
We investigated the effectiveness of the application of inbred adipose-derived stromal cells (IR-ASCs) in high inbred rat plasma (IRP) (6%)-Dulbecco modified Eagle medium (DMEM) gel with fragmin/protamine microparticles (F/P MPs) (IR-ASCs + IRP-DMEM gel + F/P MPs) on wound healing in streptozotocin-induced diabetic rats. F/P MPs have previously been used as a cell carrier for IR-ASCs in inbred Fisher 344 rats and for preservation and controlled release of various cytokines in IRP-DMEM gel. We applied IR-ASCs + IRP-DMEM gel + F/P MPs to full-thickness skin excisions on the backs of the diabetic rats. The statistical significance of wound closure was evaluated on postwounding days 3, 7, 10, and 14, and the skin area surrounding the wound was removed for histological examination on days 7 and 14. The wound closure rate and histological examination of wounds treated with IR-ASCs + IRP-DMEM gel + F/P MPs demonstrated significantly advanced epithelialization, capillary formation, and granulation tissue formation. When DiI-labeled IR-ASCs + IRP-DMEM gel + F/P MPs were applied to full-thickness skin wounds on the backs of the diabetic rats, histological observation at 2 weeks showed appearances of both DiI-labeled granulation tissue and CD31-immunostained microvessels in the transplant areas. A portion of the transplanted IR-ASCs + IRP-DMEM gel + F/P MPs had been taken up into the granulation tissues to promote wound healing. Thus, IR-ASCs + IRP-DMEM gel + F/P MPs were effective for repairing healing-impaired wounds such as those arising in the diabetic rats.
Collapse
|
42
|
Irmak G, Demirtaş TT, Çetin Altındal D, Çalış M, Gümüşderelioğlu M. Sustained Release of 17β-Estradiol Stimulates Osteogenic Differentiation of Adipose Tissue-Derived Mesenchymal Stem Cells on Chitosan-Hydroxyapatite Scaffolds. Cells Tissues Organs 2014; 199:37-50. [DOI: 10.1159/000362362] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2014] [Indexed: 11/19/2022] Open
|
43
|
Adipose-derived stromal cells for osteoarticular repair: trophic function versus stem cell activity. Expert Rev Mol Med 2014; 16:e9. [PMID: 24810570 PMCID: PMC4017835 DOI: 10.1017/erm.2014.9] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The identification of multipotent adipose-derived stromal cells (ASC) has raised hope that tissue regeneration approaches established with bone-marrow-derived stromal cells (BMSC) can be reproduced with a cell-type that is far more accessible in large quantities. Recent detailed comparisons, however, revealed subtle functional differences between ASC and BMSC, stressing the concept of a common mesenchymal progenitor existing in a perivascular niche across all tissues. Focussing on bone and cartilage repair, this review summarises recent in vitro and in vivo studies aiming towards tissue regeneration with ASC. Advantages of good accessibility, high yield and superior growth properties are counterbalanced by an inferiority of ASC to form ectopic bone and stimulate long-bone healing along with their less pronounced osteogenic and angiogenic gene expression signature. Hence, particular emphasis is placed on establishing whether stem cell activity of ASC is so far proven and relevant for successful osteochondral regeneration, or whether trophic activity may largely determine therapeutic outcome.
Collapse
|
44
|
Liang H, Li X, Shimer AL, Balian G, Shen FH. A novel strategy of spine defect repair with a degradable bioactive scaffold preloaded with adipose-derived stromal cells. Spine J 2014; 14:445-54. [PMID: 24360747 DOI: 10.1016/j.spinee.2013.09.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 09/27/2013] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Although the use of mesenchymal stem cells (MSC) with scaffolds for bone repair has been considered an effective method, the interactions between implanted materials and bone tissues have not been fully elucidated. At some specific sites, such as the vertebral body (VB) of the spine, the process of bone repair with implanted biomaterials is rarely reported. Recently, adipose tissue was found to be an alternative source of MSC besides bone marrow. However, the strategy of using adipose-derived stromal (ADS) cells with bioactive scaffold for the repair of spinal bone defects has seldom been studied. PURPOSE To use a sintered poly(lactide-co-glycolide) acid (PLGA) microspheres scaffold seeded with induced rat ADS cells to repair a bone defect of the VB in a rat model. STUDY DESIGN Basic science and laboratory study. METHODS A sintered porous microspheres scaffold was manufactured by PLGA. ADS cells were isolated from Fischer 344 rats and then induced by osteogenic medium with growth and differentiation factor 5 (GDF5) in vitro. Before implantation, cells were cultured with inductive media for 2 weeks as a monolayer situation and 1 more week on a PLGA scaffold as a three-dimensional structure. These assembled bioactive scaffolds then were implanted in lumbar VB bone defects in Fischer 344 rats. The ex vivo differentiation of the cells was confirmed by von Kossa staining and real-time polymerase chain reaction. The performance of cells on the scaffold was detected by scanning electron microscopy and (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) assay. In vivo bone formation was quantitatively measured by computed tomography study. And the effect of tissue repair was also evaluated by histological studies. RESULTS Proliferation and differentiation of cells were confirmed before in vivo implantation. Quantification of bone formation in vivo through serial three-dimensional computed tomography images revealed that the VB implanted with GDF5-induced cells demonstrated more bone formation than the control groups. Besides the bone formation period that occurred between 2 and 4 weeks in all groups, a second bone formation period was found to occur only in the groups that received cells with previous induction in vitro. This second period of significant bone formation happened simultaneously with collapsing of the scaffolds. It was then demonstrated histologically that vascularization early in the process and cooperation between host bone and implanted cells accompanied by collapse of the scaffold may be the factors that influence bone formation. This study not only provides a therapeutic strategy of using biomaterial for bone repair in the spine, but also may lead to a technological method for studying the relationship between implanted stem cells and host tissue. CONCLUSIONS Adipose-derived stromal cells maintained in culture on a scaffold and treated with osteogenic induction with growth factor ex vivo could be used to enhance bone repair in vivo.
Collapse
Affiliation(s)
- Haixiang Liang
- Department of Orthopaedic Surgery, Orthopaedic Research Laboratories, University of Virginia School of Medicine, Cobb Hall, Room B057, Box 800159, Charlottesville, VA 22908, USA
| | - Xudong Li
- Department of Orthopaedic Surgery, Orthopaedic Research Laboratories, University of Virginia School of Medicine, Cobb Hall, Room B057, Box 800159, Charlottesville, VA 22908, USA
| | - Adam L Shimer
- Department of Orthopaedic Surgery, Orthopaedic Research Laboratories, University of Virginia School of Medicine, Cobb Hall, Room B057, Box 800159, Charlottesville, VA 22908, USA
| | - Gary Balian
- Department of Orthopaedic Surgery, Orthopaedic Research Laboratories, University of Virginia School of Medicine, Cobb Hall, Room B057, Box 800159, Charlottesville, VA 22908, USA; Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Jordan Hall 6007, 1340 Jefferson Park Ave, Charlottesville, VA 22908, USA
| | - Francis H Shen
- Department of Orthopaedic Surgery, Orthopaedic Research Laboratories, University of Virginia School of Medicine, Cobb Hall, Room B057, Box 800159, Charlottesville, VA 22908, USA.
| |
Collapse
|
45
|
Effects of harvesting sites and ages on adipose tissue-derived stem cells in rat. Tissue Eng Regen Med 2014. [DOI: 10.1007/s13770-014-0410-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
46
|
Aziz Aly LA, Menoufy HE, Ragae A, Rashed LA, Sabry D. Adipose stem cells as alternatives for bone marrow mesenchymal stem cells in oral ulcer healing. Int J Stem Cells 2013; 5:104-14. [PMID: 24298363 DOI: 10.15283/ijsc.2012.5.2.104] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2012] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Adipose tissue is now recognized as an accessible, abundant, and reliable site for the isolation of adult stem cells suitable for tissue engineering and regenerative medicine applications. METHODS AND RESULTS Oral ulcers were induced by topical application of formocresol in the oral cavity of dogs. Transplantation of undifferentiated GFP-labeled Autologous Bone Marrow Stem Cell (BMSCs), Adipose Derived Stem Cell (ADSCs) or vehicle (saline) was injected around the ulcer in each group. The healing process of the ulcer was monitored clinically and histopathologically. Gene expression of vascular endothelial growth factor (VEGF) was detected in MSCs by Reverse Transcription-Polymerase Chain Reaction (RT-PCR). Expression of VEGF and collagen genes was detected in biopsies from all ulcers. RESULTS MSCs expressed mRNA for VEGF MSCs transplantation significantly accelerated oral ulcer healing compared with controls. There was increased expression of both collagen and VEGF genes in MSCs-treated ulcers compared to controls. CONCLUSIONS MSCs transplantation may help to accelerate oral ulcer healing, possibly through the induction of angiogenesis by VEGF together with increased intracellular matrix formation as detected by increased collagen gene expression. This body of work has provided evidence supporting clinical applications of adipose-derived cells in safety and efficacy trials as an alternative for bone marrow mesenchymal stem cells in oral ulcer healing.
Collapse
Affiliation(s)
- Lobna Abdel Aziz Aly
- Assistant Professor of Oral and Maxillofacial Surgery, Faculty of Dentistry, Future University
| | | | | | | | | |
Collapse
|
47
|
Sousa BR, Parreira RC, Fonseca EA, Amaya MJ, Tonelli FMP, Lacerda SMSN, Lalwani P, Santos AK, Gomes KN, Ulrich H, Kihara AH, Resende RR. Human adult stem cells from diverse origins: An overview from multiparametric immunophenotyping to clinical applications. Cytometry A 2013; 85:43-77. [DOI: 10.1002/cyto.a.22402] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 09/27/2013] [Accepted: 10/01/2013] [Indexed: 02/06/2023]
Affiliation(s)
- Bruna R. Sousa
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Ricardo C. Parreira
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Emerson A Fonseca
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Maria J. Amaya
- Department of Internal Medicine, Section of Digestive Diseases; Yale University School of Medicine; New Haven Connecticut
| | - Fernanda M. P. Tonelli
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Samyra M. S. N. Lacerda
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Pritesh Lalwani
- Faculdade de Ciências Farmacêuticas; Universidade Federal do Amazonas; Manaus AM Brazil
| | - Anderson K. Santos
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Katia N. Gomes
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Henning Ulrich
- Departamento de Bioquímica; Instituto de Química, Universidade de São Paulo; São Paulo SP Brazil
| | - Alexandre H. Kihara
- Núcleo de Cognição e Sistemas Complexos, Centro de Matemática, Computação e Cognição; Universidade Federal do ABC; Santo André SP Brazil
| | - Rodrigo R. Resende
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| |
Collapse
|
48
|
Development of a new pre-vascularized tissue-engineered construct using pre-differentiated rADSCs, arteriovenous vascular bundle and porous nano-hydroxyapatide-polyamide 66 scaffold. BMC Musculoskelet Disord 2013; 14:318. [PMID: 24209783 PMCID: PMC3826526 DOI: 10.1186/1471-2474-14-318] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2013] [Accepted: 11/05/2013] [Indexed: 01/22/2023] Open
Abstract
Background Development of a pre-vascularized tissue-engineered construct with intrinsic vascular system for cell growth and tissue formation still faces many difficulties due to the complexity of the vascular network of natural bone tissue. The present study was to design and form a new vascularized tissue-engineered construct using pre-differentiated rADSCs, arteriovenous vascular bundle and porous nHA-PA 66 scaffold. Methods rADSCs were pre-differentiated to endothelial cells (rADSCs-Endo) and then incorporated in nHA-PA 66 scaffolds in vitro. Subsequently, in vivo experiments were carried out according to the following groups: Group A (rADSCs-Endo/nHA-PA 66 scaffold with arteriovenous vascular bundle), Group B (rADSCs/nHA-PA 66 scaffold with arteriovenous vascular bundle); Group C (nHA-PA66 scaffold with arteriovenous vascular bundle), Group D (nHA-PA 66 scaffold only). The vessel density and vessel diameter were measured based on histological and immunohistochemical evaluation, furthermore, the VEGF-C, FGF-2 and BMP-2 protein expressions were also evaluated by western blot analysis. Results The results of in vivo experiments showed that the vessel density and vessel diameter in group A were significantly higher than the other three groups. Between Group B and C, no statistical difference was observed at each time point. In accordance with the results, there were dramatically higher expressions of VEGF-C and FGF-2 protein in Group A than that of Group B, C and D at 2 or 4 weeks. Statistical differences were not observed in VEGF-C and FGF-2 expression between Group B and C. BMP-2 was not expressed in any group at each time point. Conclusions Compared with muscular wrapping method, arteriovenous vascular bundle implantation could promote vascularization of the scaffold; and the angiogenesis of the scaffold was significantly accelerated when pre-differentiated rADSCs (endothelial differentiation) were added. These positive results implicate the combination of pre-differentiated rADSCs (endothelial differentiation) and arteriovenous vascular bundle may achieve rapidly angiogenesis of biomaterial scaffold.
Collapse
|
49
|
Yang P, Huang X, Wang C, Dang X, Wang K. Repair of bone defects using a new biomimetic construction fabricated by adipose-derived stem cells, collagen I, and porous beta-tricalcium phosphate scaffolds. Exp Biol Med (Maywood) 2013; 238:1331-43. [PMID: 24157587 DOI: 10.1177/1535370213505827] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Adipose derived stem cells (ASCs) with multilineage differentiation capacities have been demonstrated as an alternative cell candidate for in vitro and in vivo bone regeneration. This suggests that they may be a potential candidate to repair the bone defects. We attempted to demonstrate the use of new biomimetic constructions of undifferentiated rabbit adipose-derived stem cells (rASCs) with fully interconnected porous beta-tricalcium phosphate (β-TCP) scaffolds encapsulated by collagen I hydrogel in the regeneration of a critical-sized defect of rabbit radii. Critical-sized defects in the left radii of rabbits were prepared and inserted with rASCs/collagen I/β-TCP scaffold composites or collagen I/β-TCP scaffold composites. The results were evaluated by histology, radiographs, micro-CT, Emission Computed Tomography (ECT), fluorochrome labeling, western blot, and mechanical testing at 4, 8, and 12 weeks postsurgery. Twelve weeks after implantation, the defects were almost completely repaired as confirmed by the presence of the cortical bone and medullary cavity, which was evaluated through radiologic, histologic, and biomechanical examination. Biodegradation of the biomaterials may be attributed to extracellular liquid dissolution together with cell-mediated phagocytosis. Our study shows that a greater number of rASCs in the porous β-TCP scaffold encapsulated by collagen I gel enhanced osteogenesis in critical-sized defects. We hope to garner new insight into the engineering of rASCs-based bone tissue for clinical application.
Collapse
Affiliation(s)
- Pei Yang
- Department of Orthopaedics, the Second Affiliated Hospital of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710004, China
| | | | | | | | | |
Collapse
|
50
|
Fernandes TJ, Hodge JM, Singh PP, Eeles DG, Collier FM, Holten I, Ebeling PR, Nicholson GC, Quinn JMW. Cord blood-derived macrophage-lineage cells rapidly stimulate osteoblastic maturation in mesenchymal stem cells in a glycoprotein-130 dependent manner. PLoS One 2013; 8:e73266. [PMID: 24069182 PMCID: PMC3772005 DOI: 10.1371/journal.pone.0073266] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Accepted: 07/22/2013] [Indexed: 11/18/2022] Open
Abstract
In bone, depletion of osteoclasts reduces bone formation in vivo, as does osteal macrophage depletion. How osteoclasts and macrophages promote the action of bone forming osteoblasts is, however, unclear. Since recruitment and differentiation of multi-potential stromal cells/mesenchymal stem cells (MSC) generates new active osteoblasts, we investigated whether human osteoclasts and macrophages (generated from cord blood-derived hematopoietic progenitors) induce osteoblastic maturation in adipose tissue-derived MSC. When treated with an osteogenic stimulus (ascorbate, dexamethasone and β-glycerophosphate) these MSC form matrix-mineralising, alkaline phosphatase-expressing osteoblastic cells. Cord blood-derived progenitors were treated with macrophage colony stimulating factor (M-CSF) to form immature proliferating macrophages, or with M-CSF plus receptor activator of NFκB ligand (RANKL) to form osteoclasts; culture medium was conditioned for 3 days by these cells to study their production of osteoblastic factors. Both osteoclast- and macrophage-conditioned medium (CM) greatly enhanced MSC osteoblastic differentiation in both the presence and absence of osteogenic medium, evident by increased alkaline phosphatase levels within 4 days and increased mineralisation within 14 days. These CM effects were completely ablated by antibodies blocking gp130 or oncostatin M (OSM), and OSM was detectable in both CM. Recombinant OSM very potently stimulated osteoblastic maturation of these MSC and enhanced bone morphogenetic protein-2 (BMP-2) actions on MSC. To determine the influence of macrophage activation on this OSM-dependent activity, CM was collected from macrophage populations treated with M-CSF plus IL-4 (to induce alternative activation) or with GM-CSF, IFNγ and LPS to cause classical activation. CM from IL-4 treated macrophages stimulated osteoblastic maturation in MSC, while CM from classically-activated macrophages did not. Thus, macrophage-lineage cells, including osteoclasts but not classically activated macrophages, can strongly drive MSC-osteoblastic commitment in OSM-dependent manner. This supports the notion that eliciting gp130-dependent signals in human MSC would be a useful approach to increase bone formation.
Collapse
Affiliation(s)
- Tania J. Fernandes
- Northwest Academic Centre, Department of Medicine, The University of Melbourne, Victoria, Australia
- Barwon Biomedical Research, The Geelong Hospital, Geelong, Victoria, Australia
| | - Jason M. Hodge
- Northwest Academic Centre, Department of Medicine, The University of Melbourne, Victoria, Australia
- Barwon Biomedical Research, The Geelong Hospital, Geelong, Victoria, Australia
- School of Medicine, Deakin University: Barwon Health, Geelong, Victoria, Australia
| | | | - Damien G. Eeles
- Prince Henry's Institute of Medical Research, Clayton, Victoria, Australia
- Department of Human Biosciences, La Trobe University, Bundoora, Victoria, Australia
| | - Fiona M. Collier
- Barwon Biomedical Research, The Geelong Hospital, Geelong, Victoria, Australia
- School of Medicine, Deakin University: Barwon Health, Geelong, Victoria, Australia
| | - Ian Holten
- Department of Plastic Surgery, Barwon Health, Geelong, Victoria, Australia
| | - Peter R. Ebeling
- Northwest Academic Centre, Department of Medicine, The University of Melbourne, Victoria, Australia
| | - Geoffrey C. Nicholson
- Northwest Academic Centre, Department of Medicine, The University of Melbourne, Victoria, Australia
- Rural Clinical School, The University of Queensland, Toowoomba, Queensland, Australia
| | - Julian M. W. Quinn
- Prince Henry's Institute of Medical Research, Clayton, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
- * E-mail:
| |
Collapse
|