1
|
Zhao X, Li L, Zhang Y, Liu Z, Xing H, Gu Z. 3D Printing and Property of Biomimetic Hydroxyapatite Scaffold. Biomimetics (Basel) 2024; 9:714. [PMID: 39590286 PMCID: PMC11591832 DOI: 10.3390/biomimetics9110714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 11/09/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
The 3D printing of a biomimetic scaffold with a high hydroxyapatite (HA) content (>80%) and excellent mechanical property is a serious challenge because of the difficulty of forming and printing, insufficient cohesion, and low mechanical property of the scaffold. In this study, hydroxyapatite whiskers (HAWs), with their superior mechanical property, biodegradability, and biocompatibility, were used to reinforce spherical HA scaffolds by 3D printing. The compressive strength and energy absorption capacity of HAW-reinforced spherical HA (HAW/HA) scaffolds increased when the HAW/HA ratio increased from 0:10 to 4:6 and then dropped with any further increases in the HAW/HA ratio. Bioceramic content (HAWs and spherical HA) in the scaffolds reached 83%, and the scaffold with a HAW/HA ratio of 4:6 (4-HAW/HA) exhibited an optimum compressive strength and energy absorption capacity. The scaffold using polyvinyl alcohol (PVA) as an additive possessed a good bonding between HA and PVA as well as a higher strength, which allowed the scaffold to bear a higher stress at the same strain. The compressive strength and toughness of the 4-HAW/HA-PVA scaffold were 1.96 and 1.63 times that of the 4-HAW/HA scaffold with hydroxypropyl methyl cellulose (HPMC), respectively. The mechanical property and inorganic components of the biomimetic HAW/HA scaffold were similar to those of human bone, which would make it ideal for repairing bone defects.
Collapse
Affiliation(s)
- Xueni Zhao
- College of Mechanical and Electrical Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (L.L.)
| | | | | | | | | | | |
Collapse
|
2
|
Basile M, Marchegiani F, Novak S, Kalajzic I, Di Pietro R. Human amniotic fluid stem cells attract osteoprogenitor cells in bone healing. J Cell Physiol 2020; 235:4643-4654. [PMID: 31650536 PMCID: PMC7018542 DOI: 10.1002/jcp.29342] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 09/30/2019] [Indexed: 02/05/2023]
Abstract
Current treatments of large bone defects are based on autologous or allogenic bone transplantation. Human amniotic fluid stem cells (hAFSCs) were evaluated for their potential in bone regenerative medicine. In this study, hAFSCs were transduced with lentiviral vector harboring red fluorescent protein to investigate their role in the regeneration of critical-size bone defects in calvarial mouse model. To distinguish donor versus recipient cells, a transgenic mouse model carrying GFP fluorescent reporter was used as recipient to follow the fate of hAFSCs transplanted in vivo into Healos® scaffold. Our results showed that transduced hAFSCs can be tracked in vivo directly at the site of transplantation. The presence of GFP positive cells in the scaffold at 3 and 6 weeks after transplantation indicates that donor hAFSCs can recruit host cells during the repair process. These observations help clarify the role of hAFSCs in bone tissue repair.
Collapse
Affiliation(s)
- Mariangela Basile
- Department of Reconstructive Sciences, UConn Health, Farmington, Connecticut
- Department of Medicine and Ageing Sciences, G. d’Annunzio University of Chieti-Pescara, Italy
- StemTeCh Group, CAST, G. d’Annunzio University of Chieti-Pescara, Italy
| | - Francesco Marchegiani
- Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, Padua, Italy
| | - Sanja Novak
- Department of Reconstructive Sciences, UConn Health, Farmington, Connecticut
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, UConn Health, Farmington, Connecticut
| | - Roberta Di Pietro
- Department of Medicine and Ageing Sciences, G. d’Annunzio University of Chieti-Pescara, Italy
- StemTeCh Group, CAST, G. d’Annunzio University of Chieti-Pescara, Italy
| |
Collapse
|
3
|
FERNÁNDEZ MPEÑA, WITTE F, TOZZI G. Applications of X‐ray computed tomography for the evaluation of biomaterial‐mediated bone regeneration in critical‐sized defects. J Microsc 2020; 277:179-196. [DOI: 10.1111/jmi.12844] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 10/06/2019] [Accepted: 11/04/2019] [Indexed: 12/16/2022]
Affiliation(s)
- M. PEÑA FERNÁNDEZ
- Zeiss Global Centre, School of Mechanical and Design EngineeringUniversity of Portsmouth Portsmouth UK
| | - F. WITTE
- Biotrics Bioimplants GmbH Berlin Germany
| | - G. TOZZI
- Zeiss Global Centre, School of Mechanical and Design EngineeringUniversity of Portsmouth Portsmouth UK
| |
Collapse
|
4
|
Mohammed EEA, Beherei HH, El-Zawahry M, Farrag ARH, Kholoussi N, Helwa I, Gaber K, Allam MA, Mabrouk M, Aleem AKA. Combination of Human Amniotic Fluid Derived-Mesenchymal Stem Cells and Nano-hydroxyapatite Scaffold Enhances Bone Regeneration. Open Access Maced J Med Sci 2019; 7:2739-2750. [PMID: 31844430 PMCID: PMC6901872 DOI: 10.3889/oamjms.2019.730] [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: 08/06/2019] [Revised: 08/29/2019] [Accepted: 08/30/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND: Human amniotic fluid-derived stem cells (hAF-MSCs) have a high proliferative capacity and osteogenic differentiation potential in vitro. The combination of hAF-MSCs with three-dimensional (3D) scaffold has a promising therapeutic potential in bone tissue engineering and regenerative medicine. Selection of an appropriate scaffold material has a crucial role in a cell supporting and osteoinductivity to induce new bone formation in vivo. AIM: This study aimed to investigate and evaluate the osteogenic potential of the 2nd-trimester hAF-MSCs in combination with the 3D scaffold, 30% Nano-hydroxyapatite chitosan, as a therapeutic application for bone healing in the induced tibia defect in the rabbit. SUBJECT AND METHODS: hAF-MSCs proliferation and culture expansion was done in vitro, and osteogenic differentiation characterisation was performed by Alizarin Red staining after 14 & 28 days. Expression of the surface markers of hAF-MSCs was assessed using Flow Cytometer with the following fluorescein-labelled antibodies: CD34-PE, CD73-APC, CD90-FITC, and HLA-DR-FITC. Ten rabbits were used as an animal model with an induced defect in the tibia to evaluate the therapeutic potential of osteogenic differentiation of hAF-MSCs seeded on 3D scaffold, 30% Nano-hydroxyapatite chitosan. The osteogenic differentiated hAF-MSCs/scaffold composite system applied and fitted in the defect region and non-seeded scaffold was used as control. The histopathological investigation was performed at 2, 3, & 4 weeks post-transplantation and scanning electron microscope (SEM) was assessed at 2 & 4 weeks post-transplantation to evaluate the bone healing potential in the rabbit tibia defect. RESULTS: Culture and expansion of 2nd-trimester hAF-MSCs presented high proliferative and osteogenic potential in vitro. Histopathological examination for the transplanted hAF-MSCs seeded on the 3D scaffold, 30% Nano-hydroxyapatite chitosan, demonstrated new bone formation in the defect site at 2 & 3 weeks post-transplantation as compared to the control (non-seeded scaffold). Interestingly, the scaffold accelerated the osteogenic differentiation of AF-MSCs and showed complete bone healing of the defect site as compared to the control (non-seeded scaffold) at 4 weeks post-transplantation. Furthermore, the SEM analysis confirmed these findings. CONCLUSION: The combination of the 2nd-trimester hAF-MSCs and 3D scaffold, 30% Nano-hydroxyapatite chitosan, have a therapeutic perspective for large bone defect and could be used effectively in bone tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Eman E A Mohammed
- Medical Molecular Genetics Department, National Research Centre, Cairo, Egypt.,Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, Cairo, Egypt
| | - Hanan H Beherei
- Ceramic Department (Biomaterials), National Research Centre, Cairo, Egypt
| | - Mohamed El-Zawahry
- Fixed and Removable Prosthodontics Department, National Research Centre, Cairo, Egypt
| | | | - Naglaa Kholoussi
- Immunogenetics Department, National Research Centre, Cairo, Egypt
| | - Iman Helwa
- Immunogenetics Department, National Research Centre, Cairo, Egypt
| | - Khaled Gaber
- Prenatal and Fetal medicine Department, National Research Centre, Cairo, Egypt
| | - Mousa A Allam
- Spectroscopy Department, National Research Centre, Cairo, Egypt
| | - Mostafa Mabrouk
- Ceramic Department (Biomaterials), National Research Centre, Cairo, Egypt
| | - Alice K Abdel Aleem
- Medical Molecular Genetics Department, National Research Centre, Cairo, Egypt.,Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, Cairo, Egypt.,Neurology and Neuroscience Department, Weill Cornell Medical College, Doha, Qatar
| |
Collapse
|
5
|
Mohammed EEA, El-Zawahry M, Farrag ARH, Aziz NNA, Sharaf-ElDin W, Abu-Shahba N, Mahmoud M, Gaber K, Ismail T, Mossaad MM, Aleem AKA. Osteogenic Differentiation Potential of Human Bone Marrow and Amniotic Fluid-Derived Mesenchymal Stem Cells in Vitro & in Vivo. Open Access Maced J Med Sci 2019; 7:507-515. [PMID: 30894903 PMCID: PMC6420942 DOI: 10.3889/oamjms.2019.124] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/13/2019] [Accepted: 01/14/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Cell therapies offer a promising potential in promoting bone regeneration. Stem cell therapy presents attractive care modality in treating degenerative conditions or tissue injuries. The rationale behind this is both the expansion potential of stem cells into a large cell population size and its differentiation abilities into a wide variety of tissue types, when given the proper stimuli. A progenitor stem cell is a promising source of cell therapy in regenerative medicine and bone tissue engineering. AIM This study aimed to compare the osteogenic differentiation and regenerative potentials of human mesenchymal stem cells derived from human bone marrow (hBM-MSCs) or amniotic fluid (hAF-MSCs), both in vitro and in vivo studies. SUBJECTS AND METHODS Human MSCs, used in this study, were successfully isolated from two human sources; the bone marrow (BM) and amniotic fluid (AF) collected at the gestational ages of second or third trimesters. RESULTS The stem cells derived from amniotic fluid seemed to be the most promising type of progenitor cells for clinical applications. In a pre-clinical experiment, attempting to explore the therapeutic application of MSCs in bone regeneration, Rat lumbar spines defects were surgically created and treated with undifferentiated and osteogenically differentiated MSCs, derived from BM and second trimester AF. Cells were loaded on gel-foam scaffolds, inserted and fixed in the area of the surgical defect. X-Ray radiography follows up, and histopathological analysis was done three-four months post- operation. The transplantation of AF-MSCs or BM-MSCs into induced bony defects showed promising results. The AF-MSCs are offering a better healing effect increasing the likelihood of achieving successful spinal fusion. Some bone changes were observed in rats transplanted with osteoblasts differentiated cells but not in rats transplanted with undifferentiated MSCs. Longer observational periods are required to evaluate a true bone formation. The findings of this study suggested that the different sources; hBM-MSCs or hAF-MSCs exhibited remarkably different signature regarding the cell morphology, proliferation capacity and osteogenic differentiation potential. CONCLUSIONS AF-MSCs have a better performance in vivo bone healing than that of BM-MSCs. Hence, AF derived MSCs is highly recommended as an alternative source to BM-MSCs in bone regeneration and spine fusion surgeries. Moreover, the usage of gel-foam as a scaffold proved as an efficient cell carrier that showed bio-compatibility with cells, bio-degradability and osteoinductivity in vivo.
Collapse
Affiliation(s)
- Eman E A Mohammed
- Medical Molecular Genetics Department, Human Genetics and Genome Research Division, National Research Center, Cairo, Egypt.,Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, Cairo, Egypt
| | - Mohamed El-Zawahry
- Fixed and Removable Prosthodontics Department, National Research Center, Cairo, Egypt
| | - Abdel Razik H Farrag
- Pathology Department, Medical Research Division, National Research Center, Cairo, Egypt
| | - Nahla N Abdel Aziz
- Medical Molecular Genetics Department, Human Genetics and Genome Research Division, National Research Center, Cairo, Egypt
| | - Wessam Sharaf-ElDin
- Medical Molecular Genetics Department, Human Genetics and Genome Research Division, National Research Center, Cairo, Egypt
| | - Nourhan Abu-Shahba
- Medical Molecular Genetics Department, Human Genetics and Genome Research Division, National Research Center, Cairo, Egypt.,Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, Cairo, Egypt
| | - Marwa Mahmoud
- Medical Molecular Genetics Department, Human Genetics and Genome Research Division, National Research Center, Cairo, Egypt.,Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, Cairo, Egypt
| | - Khaled Gaber
- Prenatal and Fetal medicine Department, Oral and Dental Research Division, National Research Center, Cairo, Egypt
| | - Taher Ismail
- Gynecology and Obstetric Department, Faculty of Medicine (Boys), Al - Azhar University, Cairo, Egypt
| | | | - Alice K Abdel Aleem
- Medical Molecular Genetics Department, Human Genetics and Genome Research Division, National Research Center, Cairo, Egypt.,Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, Cairo, Egypt.,Neurology and Neuroscience Department, Weill Cornell Medicine Qatar, Doha, Qatar
| |
Collapse
|
6
|
Irreversible Electroporation in Liver Cancers and Whole Organ Engineering. J Clin Med 2018; 8:jcm8010022. [PMID: 30585195 PMCID: PMC6352021 DOI: 10.3390/jcm8010022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 12/15/2018] [Accepted: 12/19/2018] [Indexed: 12/18/2022] Open
Abstract
Liver cancers contribute significantly to cancer-related mortality worldwide and liver transplants remain the cornerstone of curative treatment for select, early-stage patients. Unfortunately, because of a mismatch between demand and supply of donor organs, liver cancer patients must often wait extended periods of time prior to transplant. A variety of local therapies including surgical resection, transarterial chemoembolization, and thermal ablative methods exist in order to bridge to transplant. In recent years, a number of studies have examined the role of irreversible electroporation (IRE) as a non-thermal local ablative method for liver tumors, particularly for those adjacent to critical structures such as the vasculature, gall bladder, or bile duct. In addition to proving its utility as a local treatment modality, IRE has also demonstrated promise as a technique for donor organ decellularization in the context of whole-organ engineering. Through complete non-thermal removal of living cells, IRE allows for the creation of an acellular extra cellular matrix (ECM) scaffold that could theoretically be recellularized and implanted into a living host. Here, we comprehensively review studies investigating IRE, its role in liver cancer treatment, and its utility in whole organ engineering.
Collapse
|
7
|
Loukogeorgakis SP, De Coppi P. Concise Review: Amniotic Fluid Stem Cells: The Known, the Unknown, and Potential Regenerative Medicine Applications. Stem Cells 2018; 35:1663-1673. [PMID: 28009066 DOI: 10.1002/stem.2553] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 09/07/2016] [Accepted: 10/01/2016] [Indexed: 12/19/2022]
Abstract
The amniotic fluid has been identified as an untapped source of cells with broad potential, which possess immunomodulatory properties and do not have the ethical and legal limitations of embryonic stem cells. CD117(c-Kit)+ cells selected from amniotic fluid have been shown to differentiate into cell lineages representing all three embryonic germ layers without generating tumors, making them ideal candidates for regenerative medicine applications. Moreover, their ability to engraft in injured organs and modulate immune and repair responses of host tissues, suggest that transplantation of such cells may be useful for the treatment of various degenerative and inflammatory diseases. Although significant questions remain regarding the origin, heterogeneous phenotype, and expansion potential of amniotic fluid stem cells, evidence to date supports their potential role as a valuable stem cell source for the field of regenerative medicine. Stem Cells 2017;35:1663-1673.
Collapse
Affiliation(s)
- Stavros P Loukogeorgakis
- Stem Cells and Regenerative Medicine Section, Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom
| | - Paolo De Coppi
- Stem Cells and Regenerative Medicine Section, Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
8
|
Pei W, Lu T, Wang K, Ji M, Zhang S, Chen F, Li L, Li X, Guan W. Biological characterization and pluripotent identification of ovine amniotic fluid stem cells. Cytotechnology 2018; 70:1009-1021. [PMID: 29502286 DOI: 10.1007/s10616-017-0115-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 03/21/2017] [Indexed: 01/03/2023] Open
Abstract
Mesenchymal stem cells derived from amniotic fluid have become one of the most potential stem cell source for cell-based therapy for the reason they can be harvested at low cost and without ethical problems. Here, we obtained amniotic fluid stem cells (AFSCs) from ovine amniotic fluid and studied the expansion capacity, cell markers expression, karyotype, and multilineage differentiation ability. In our work, AFSCs were subcultured to passage 62. The cell markers, CD29, CD44, CD73 and OCT4 which analyzed by RT-PCR were positive; CD44, CD73, CD90, CD105, NANOG, OCT4 analyzed by immunofluorescence and flow cytometry were also positive. The growth curves of different passages were all typically sigmoidal. The different passages cells took on a normal karyotype. In addition, AFSCs were successfully induced to differentiate into adipocytes, osteoblasts and chondrocytes. The results suggested that the AFSCs isolated from ovine maintained normal biological characteristics and their multilineage differentiation potential provides many potential applications in cell-based therapies and tissue engineering.
Collapse
Affiliation(s)
- Wenhua Pei
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West road, haidian district, Beijing, 100193, China
| | - Tengfei Lu
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West road, haidian district, Beijing, 100193, China
| | - Kunfu Wang
- College of Wildlife Resources, Northeast Forestry University, Harbin, 150040, China
| | - Meng Ji
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West road, haidian district, Beijing, 100193, China
| | - Shuang Zhang
- Research Center for Sports Scientific Experiment, Harbin Sport University, Harbin, People's Republic of China
| | - Fenghao Chen
- College of Human Movement Science, Harbin Sport University, Harbin, 150040, China
| | - Lu Li
- College of Animal Science and Technology, Agricultural University of Hebei, Baoding, 071000, China
| | - Xiangchen Li
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West road, haidian district, Beijing, 100193, China.
| | - Weijun Guan
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West road, haidian district, Beijing, 100193, China.
| |
Collapse
|
9
|
Comparative study on characterization and wound healing potential of goat (Capra hircus) mesenchymal stem cells derived from fetal origin amniotic fluid and adult bone marrow. Res Vet Sci 2017; 112:81-88. [PMID: 28135618 DOI: 10.1016/j.rvsc.2016.12.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 12/25/2016] [Accepted: 12/30/2016] [Indexed: 01/09/2023]
Abstract
Caprine amniotic fluid (cAF) and bone marrow cells (cBM) were isolated, expanded and phenotypically characterized by mesenchymal stem cells (MSCs) specific cell surface markers. Both cell types were compared for multilineage differentiation potential by flow cytometry using specific antibodies against lineage specific markers. Furthermore, in vitro expanded cAF-MSCs showed higher expression of trophic factors viz. VEGF and TGF-β1 as compared to cBM-MSCs. Full-skin thickness excisional wounds created on either side of the dorsal midline (thoracolumbar) of New Zealand White rabbits were randomly assigned to subcutaneous injection of either fetal origin cAF-MSCs (n=4) or adult cBM-MSCs (n=4) or sterile PBS (control, n=4). The rate of wound closure was found faster (p<0.05) in cAF-MSCs treated wounds as compared with cBM-MSCs and PBS treated wounds especially on 21st day post-skin excision. Histomorphological examination of the healing tissue showed that wound healing was improved (p<0.05) by greater epithelialization, neovascularization and collagen development in cAF-MSCs as compared to cBM-MSCs and PBS treated wounds.
Collapse
|
10
|
Counteracting bone fragility with human amniotic mesenchymal stem cells. Sci Rep 2016; 6:39656. [PMID: 27995994 PMCID: PMC5171815 DOI: 10.1038/srep39656] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 11/24/2016] [Indexed: 02/08/2023] Open
Abstract
The impaired maturation of bone-forming osteoblasts results in reduced bone formation and subsequent bone weakening, which leads to a number of conditions such as osteogenesis imperfecta (OI). Transplantation of human fetal mesenchymal stem cells has been proposed as skeletal anabolic therapy to enhance bone formation, but the mechanisms underlying the contribution of the donor cells to bone health are poorly understood and require further elucidation. Here, we show that intraperitoneal injection of human amniotic mesenchymal stem cells (AFSCs) into a mouse model of OI (oim mice) reduced fracture susceptibility, increased bone strength, improved bone quality and micro-architecture, normalised bone remodelling and reduced TNFα and TGFβ sigalling. Donor cells engrafted into bones and differentiated into osteoblasts but importantly, also promoted endogenous osteogenesis and the maturation of resident osteoblasts. Together, these findings identify AFSC transplantation as a countermeasure to bone fragility. These data have wider implications for bone health and fracture reduction.
Collapse
|
11
|
Zavatti M, Guida M, Maraldi T, Beretti F, Bertoni L, La Sala GB, De Pol A. Estrogen receptor signaling in the ferutinin-induced osteoblastic differentiation of human amniotic fluid stem cells. Life Sci 2016; 164:15-22. [PMID: 27629493 DOI: 10.1016/j.lfs.2016.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 09/01/2016] [Accepted: 09/10/2016] [Indexed: 12/16/2022]
Abstract
AIMS Ferutinin is a diaucane sesquiterpene with a high estrogenic activity. Since ferutinin is able to enhance osteoblastic differentiation of human amniotic fluid stem cells (hAFSCs), the aim of this study was to evaluate the role of the estrogen receptors α (ERα) and G-protein coupled receptor 30 (GPR30) in ferutinin-mediated osteoblastic differentiation. Moreover, it was investigated if MEK/ERK and PI3K/Akt signaling pathways are involved in ferutinin-induced effects. MAIN METHODS hAFSCs were cultured in a standard medium or in an osteoblastic medium for 14 or 21days and ferutinin was added at 10-8M. Immunofluorescence techniques and Western-blot 21analysis were used to study estrogen receptors and signaling pathways. KEY FINDINGS In both undifferentiated and differentiated hAFSCs we identified ERα and GPR30 with a nuclear or cytoplasmatic localization, respectively. The presence of ferutinin in the osteoblastic medium leads to an increase in ERα expression. To dissect the role of estrogen receptors, MPP and G15 were used to selectively block ERα and GPR30, respectively. Notably, ferutinin enhanced osteoblastic differentiation in cells challenged with G15. Ferutinin was able to increase ERK and Akt phosphorylations with a different timing activation. These phosphorylations were antagonized by PD0325901, a MEK inhibitor, and wortmannin, a PI3K inhibitor. Both MPP and G15 inhibited the ferutinin-induced MEK/ERK and PI3K/Akt pathway activations. In the osteoblastic condition, PD0325901, but not wortmannin, reduced the expression of OPN and RUNX-2, whereas ferutinin abrogated the down-modulation triggered by PD0325901. SIGNIFICANCE PI3K/Akt pathways seems to mediate the enhancement of hAFSCs osteoblastic differentiation triggered by ferutinin through ERα.
Collapse
Affiliation(s)
- M Zavatti
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplants, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy.
| | - M Guida
- EURAC Research, Center for Biomedicine, Bolzano, Italy
| | - T Maraldi
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplants, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - F Beretti
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplants, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - L Bertoni
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplants, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - G B La Sala
- Unit of Obstetrics and Ginecology, IRCCS-Arcispedale Santa Maria Nuova, Reggio Emilia, Italy
| | - A De Pol
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplants, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
12
|
Yanagawa F, Sugiura S, Kanamori T. Hydrogel microfabrication technology toward three dimensional tissue engineering. Regen Ther 2016; 3:45-57. [PMID: 31245472 PMCID: PMC6581842 DOI: 10.1016/j.reth.2016.02.007] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/15/2016] [Accepted: 02/18/2016] [Indexed: 02/07/2023] Open
Abstract
The development of biologically relevant three-dimensional (3D) tissue constructs is essential for the alternative methods of organ transplantation in regenerative medicine, as well as the development of improved drug discovery assays. Recent technological advances in hydrogel microfabrication, such as micromolding, 3D bioprinting, photolithography, and stereolithography, have led to the production of 3D tissue constructs that exhibit biological functions with precise 3D microstructures. Furthermore, microfluidics technology has enabled the development of the perfusion culture of 3D tissue constructs with vascular networks. In this review, we present these hydrogel microfabrication technologies for the in vitro reconstruction and cultivation of 3D tissues. Additionally, we discuss current challenges and future perspectives of 3D tissue engineering.
Collapse
Affiliation(s)
- Fumiki Yanagawa
- Drug Assay Device Research Group, Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5th, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Shinji Sugiura
- Drug Assay Device Research Group, Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5th, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Toshiyuki Kanamori
- Drug Assay Device Research Group, Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5th, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| |
Collapse
|
13
|
Glial cell line-derived neurotrophic factor induced the differentiation of amniotic fluid-derived stem cells into vascular endothelial-like cells in vitro. J Mol Histol 2015; 47:9-19. [PMID: 26712153 DOI: 10.1007/s10735-015-9649-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 12/18/2015] [Indexed: 10/22/2022]
Abstract
Amniotic fluid-derived stem cells (AFSCs) are a novel source of stem cells that are isolated and cultured from second trimester amniocentesis. Glial cell line-derived neurotrophic factor (GDNF) acts as a tissue morphogen and regulates stem cell proliferation and differentiation. This study investigated the effect of an adenovirus-mediated GDNF gene, which was engineered into AFSCs, on the cells' biological properties and whether GDNF in combination with AFSCs can be directionally differentiated into vascular endothelial-like cells in vitro. AFSCs were isolated and cultured using the plastic adherence method in vitro and identified by the transcription factor Oct-4, which is the primary marker of pluripotent stem cells. AFSCs were efficiently transfected by a GFP-labeled plasmid system of an adenovirus vector carrying the GDNF gene (Ad-GDNF-GFP). Transfected AFSCs stably expressed GDNF. Transfected AFSCs were cultured in endothelial growth medium-2 containing vascular endothelial growth factor. After 1 week, AFSCs were positive for von Willebrand factor (vWF) and CD31, which are markers of endothelial cells, and the recombinant GDNF group was significantly higher than undifferentiated controls and the GFP only group. These results demonstrated that AFSCs differentiated into vascular endothelial-like cells in vitro, and recombinant GDNF promoted differentiation. The differentiation-induced AFSCs may be used as seed cells to provide a new manner of cell and gene therapies for transplantation into the vascular injury site to promote angiogenesis.
Collapse
|
14
|
Pipino C, Di Tomo P, Mandatori D, Cianci E, Lanuti P, Cutrona MB, Penolazzi L, Pierdomenico L, Lambertini E, Antonucci I, Sirolli V, Bonomini M, Romano M, Piva R, Marchisio M, Pandolfi A. Calcium sensing receptor activation by calcimimetic R-568 in human amniotic fluid mesenchymal stem cells: correlation with osteogenic differentiation. Stem Cells Dev 2015; 23:2959-71. [PMID: 25036254 DOI: 10.1089/scd.2013.0627] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Human amniotic fluid mesenchymal stem cells (hAFMSCs) are promising for therapeutic applications in bone damage. Calcium sensing receptor (CaSR), a G protein-coupled receptor, plays a physiological role in the regulation of bone metabolism. Thus, the bone CaSR could be targeted by calcimimetic agonists, which may be potentially helpful in treating bone diseases. The aim of our study was to characterize CaSR expression in hAFMSCs and to assess the activity of calcimimetic R-568 during in vitro osteogenesis. Using western blotting, immunofluorescence, and flow cytometry, we consistently observed constitutive CaSR in osteo-differentiating hAFMSCs. Notably, both R-568 and calcium significantly enhanced hAFMSC osteogenic differentiation after exposure to osteogenic medium. To provide further evidence of the involvement of CaSR in osteogenesis, we correlated its expression with that of established osteogenic markers, that is, alkaline phosphatase (ALP), runt-related transcription factor 2 (Runx2), and osteopontin (OPN), and novel, not yet completely defined regulators of osteogenesis. Among these are β-catenin and Slug, which are mediators of Wnt signaling, and nuclear factor of activated T cells c1 (NFATc1), which plays a critical role in calcium/calcineurin signaling. Taken together, our results demonstrate that CaSR is expressed in hAFMSCs, positively correlates with osteogenic markers, and is activated by R-568. Notably, downregulation of CaSR by RNA interference supports the conclusion that CaSR activation plays a central role in hAFMSC osteogenesis. Thus, this study provides significant information on the mechanisms of hAFMSC osteogenesis, which could provide additional molecular basis for the use of calcimimetics in bone regenerative medicine.
Collapse
Affiliation(s)
- Caterina Pipino
- 1 Department of Experimental and Clinical Sciences, School of Medicine and Health Sciences, "G. d'Annunzio" University Chieti-Pescara , Chieti, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Stem cells from amniotic fluid--Potential for regenerative medicine. Best Pract Res Clin Obstet Gynaecol 2015; 31:45-57. [PMID: 26542929 DOI: 10.1016/j.bpobgyn.2015.08.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 08/31/2015] [Indexed: 12/31/2022]
Abstract
Regenerative medicine has recently been established as an emerging field focussing on repair, replacement or regeneration of cells, tissues and whole organs. The significant recent advances in the field have intensified the search for novel sources of stem cells with potential for therapy. Recently, researchers have identified the amniotic fluid as an untapped source of stem cells that are multipotent, possess immunomodulatory properties and do not have the ethical and legal limitations of embryonic stem cells. Stem cells from the amniotic fluid have been shown to differentiate into cell lineages representing all three embryonic germ layers without generating tumours, which make them an ideal candidate for tissue engineering applications. In addition, their ability to engraft in injured organs and modulate immune and repair responses of host tissues suggest that transplantation of such cells may be useful for the treatment of various degenerative and inflammatory diseases affecting major tissues/organs. This review summarises the evidence on amniotic fluid cells over the past 15 years and explores the potential therapeutic applications of amniotic fluid stem cells and amniotic fluid mesenchymal stem cells.
Collapse
|
16
|
Gładysz D, Hozyasz KK. Stem cell regenerative therapy in alveolar cleft reconstruction. Arch Oral Biol 2015; 60:1517-32. [PMID: 26263541 DOI: 10.1016/j.archoralbio.2015.07.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 05/23/2015] [Accepted: 07/04/2015] [Indexed: 12/17/2022]
Abstract
Achieving a successful and well-functioning reconstruction of craniofacial deformities still remains a challenge. As for now, autologous bone grafting remains the gold standard for alveolar cleft reconstruction. However, its aesthetic and functional results often remain unsatisfactory, which carries a long-term psychosocial and medical sequelae. Therefore, searching for novel therapeutic approaches is strongly indicated. With the recent advances in stem cell research, cell-based tissue engineering strategies move from the bench to the patients' bedside. Successful stem cell engineering employs a carefully selected stem cell source, a biodegradable scaffold with osteoconductive and osteoinductive properties, as well as an addition of growth factors or cytokines to enhance osteogenesis. This review highlights recent advances in mesenchymal stem cell tissue engineering, discusses animal models and case reports of stem cell enhanced bone regeneration, as well as ongoing clinical trials.
Collapse
Affiliation(s)
- Dominika Gładysz
- Department of Pediatrics, Institute of Mother and Child, Warsaw, Poland
| | - Kamil K Hozyasz
- Department of Pediatrics, Institute of Mother and Child, Warsaw, Poland.
| |
Collapse
|
17
|
Effect of Passage, Sorting, and Media on Differentiation Capacity and Marker Expression in Amniotic Fluid Stem Cells. Cell Mol Bioeng 2015. [DOI: 10.1007/s12195-015-0407-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
18
|
Pipino C, Pandolfi A. Osteogenic differentiation of amniotic fluid mesenchymal stromal cells and their bone regeneration potential. World J Stem Cells 2015; 7:681-690. [PMID: 26029340 PMCID: PMC4444609 DOI: 10.4252/wjsc.v7.i4.681] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 01/16/2015] [Accepted: 02/11/2015] [Indexed: 02/06/2023] Open
Abstract
In orthopedics, tissue engineering approach using stem cells is a valid line of treatment for patients with bone defects. In this context, mesenchymal stromal cells of various origins have been extensively studied and continue to be a matter of debate. Although mesenchymal stromal cells from bone marrow are already clinically applied, recent evidence suggests that one may use mesenchymal stromal cells from extra-embryonic tissues, such as amniotic fluid, as an innovative and advantageous resource for bone regeneration. The use of cells from amniotic fluid does not raise ethical problems and provides a sufficient number of cells without invasive procedures. Furthermore, they do not develop into teratomas when transplanted, a consequence observed with pluripotent stem cells. In addition, their multipotent differentiation ability, low immunogenicity, and anti-inflammatory properties make them ideal candidates for bone regenerative medicine. We here present an overview of the features of amniotic fluid mesenchymal stromal cells and their potential in the osteogenic differentiation process. We have examined the papers actually available on this regard, with particular interest in the strategies applied to improve in vitro osteogenesis. Importantly, a detailed understanding of the behavior of amniotic fluid mesenchymal stromal cells and their osteogenic ability is desirable considering a feasible application in bone regenerative medicine.
Collapse
|
19
|
Si JW, Wang XD, Shen SGF. Perinatal stem cells: A promising cell resource for tissue engineering of craniofacial bone. World J Stem Cells 2015; 7:149-159. [PMID: 25621114 PMCID: PMC4300925 DOI: 10.4252/wjsc.v7.i1.149] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 08/28/2014] [Accepted: 09/17/2014] [Indexed: 02/06/2023] Open
Abstract
In facing the mounting clinical challenge and suboptimal techniques of craniofacial bone defects resulting from various conditions, such as congenital malformations, osteomyelitis, trauma and tumor resection, the ongoing research of regenerative medicine using stem cells and concurrent advancement in biotechnology have shifted the focus from surgical reconstruction to a novel stem cell-based tissue engineering strategy for customized and functional craniofacial bone regeneration. Given the unique ontogenetical and cell biological properties of perinatal stem cells, emerging evidence has suggested these extraembryonic tissue-derived stem cells to be a promising cell source for extensive use in regenerative medicine and tissue engineering. In this review, we summarize the current achievements and obstacles in stem cell-based craniofacial bone regeneration and subsequently we address the characteristics of various types of perinatal stem cells and their novel application in tissue engineering of craniofacial bone. We propose the promising feasibility and scope of perinatal stem cell-based craniofacial bone tissue engineering for future clinical application.
Collapse
|
20
|
Boerckel JD, Mason DE, McDermott AM, Alsberg E. Microcomputed tomography: approaches and applications in bioengineering. Stem Cell Res Ther 2014; 5:144. [PMID: 25689288 PMCID: PMC4290379 DOI: 10.1186/scrt534] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Microcomputed tomography (microCT) has become a standard and essential tool for quantifying structure-function relationships, disease progression, and regeneration in preclinical models and has facilitated numerous scientific and bioengineering advancements over the past 30 years. In this article, we recount the early events that led to the initial development of microCT and review microCT approaches for quantitative evaluation of bone, cartilage, and cardiovascular structures, with applications in fundamental structure-function analysis, disease, tissue engineering, and numerical modeling. Finally, we address several next-generation approaches under active investigation to improve spatial resolution, acquisition time, tissue contrast, radiation dose, and functional and molecular information.
Collapse
|
21
|
Bajek A, Olkowska J, Gurtowska N, Kloskowski T, Walentowicz-Sadlecka M, Sadlecki P, Grabiec M, Drewa T. Human amniotic-fluid-derived stem cells: a unique source for regenerative medicine. Expert Opin Biol Ther 2014; 14:831-9. [PMID: 24655038 DOI: 10.1517/14712598.2014.898749] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION The first application of tissue engineering was based on the use of differentiated cells from the adult organism, which was associated with an invasiveness and high risk of diseased cells' transplantation. Over the years, the range of available cell populations for tissue engineering has widened. AREAS COVERED We review the comprehensive information concerning the characteristic features of amniotic-fluid-derived stem cells (AFSCs). We also review the potential applications of these cells in clinical practice. EXPERT OPINION AFSCs hold promise for the future treatment of many incurable diseases. However, such cell-based therapies have some limitations, and there are questions relating to the use of stem cells, which should be carefully analyzed before translation of these cells into clinical practice.
Collapse
Affiliation(s)
- Anna Bajek
- Nicolaus Copernicus University, Department of Tissue Engineering , Karlowicza 24, 85-092 Bydgoszcz , Poland +48 525853737 ; +48 525853742 ;
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Kang KS, Hong JM, Jeong YH, Seol YJ, Yong WJ, Rhie JW, Cho DW. Combined effect of three types of biophysical stimuli for bone regeneration. Tissue Eng Part A 2014; 20:1767-77. [PMID: 24446961 DOI: 10.1089/ten.tea.2013.0157] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Pretreatment using various types of biophysical stimuli could provide appropriate potential to cells during construction of the engineered tissue in vitro. We hypothesized that multiple combinations of these biophysical stimuli could enhance osteogenic differentiation in vitro and bone formation in vivo. Cyclic strain, an electromagnetic field, and ultrasound were selected and combined as effective stimuli for osteogenic differentiation using a developed bioreactor. Here we report the experimental evaluation of the osteogenic effects of various combinations of three different biophysical stimuli in vitro and in vivo using human adipose-derived stem cells (ASCs). Osteogenic differentiation of ASCs was accelerated by multiple-combination biophysical stimulation in vitro. However, both single stimulation and double-combination stimulation were sufficient to accelerate bone regeneration in vivo, while the osteogenic marker expression of those groups was not as high as that of triple-combination stimulation in vitro. We inferred from these data that ASCs appropriately differentiated into the osteogenic lineage by biophysical stimulation could be a better option for accelerating bone formation in vivo than relatively undifferentiated or completely differentiated ASCs. Although many questions remain about the mechanisms of combined effects of various biophysical stimuli, this approach could be a more powerful tool for bone tissue regeneration.
Collapse
Affiliation(s)
- Kyung Shin Kang
- 1 Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH) , Pohang, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Significant advances in the field of regenerative medicine have intensified the search for novel sources of stem cells with potential for therapy. Although embryonic and adult tissues can be used for the isolation of pluripotent stem cells, significant limitations including ethical concerns, complexity of isolation/culture and tumorigenicity have hindered translation of laboratory findings to clinical practice.
Collapse
|
24
|
Mirabella T, Gentili C, Daga A, Cancedda R. Amniotic fluid stem cells in a bone microenvironment: Driving host angiogenic response. Stem Cell Res 2013; 11:540-51. [DOI: 10.1016/j.scr.2013.02.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 02/10/2013] [Accepted: 02/11/2013] [Indexed: 12/17/2022] Open
|
25
|
Chen J, Dosier CR, Park JH, De S, Guldberg RE, Boyan BD, Schwartz Z. Mineralization of three-dimensional osteoblast cultures is enhanced by the interaction of 1α,25-dihydroxyvitamin D3 and BMP2 via two specific vitamin D receptors. J Tissue Eng Regen Med 2013; 10:40-51. [DOI: 10.1002/term.1770] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 04/16/2013] [Indexed: 11/08/2022]
Affiliation(s)
- Jiaxuan Chen
- Wallace H. Coulter Department of Biomedical Engineering; Georgia Institute of Technology; Atlanta GA USA
| | - Christopher R. Dosier
- Woodruff School of Mechanical Engineering; Georgia Institute of Technology; Atlanta GA USA
| | - Jung Hwa Park
- School of Materials Science and Engineering; Georgia Institute of Technology; Atlanta GA USA
| | - Subhendu De
- Wallace H. Coulter Department of Biomedical Engineering; Georgia Institute of Technology; Atlanta GA USA
| | - Robert E. Guldberg
- Woodruff School of Mechanical Engineering; Georgia Institute of Technology; Atlanta GA USA
| | - Barbara D. Boyan
- Wallace H. Coulter Department of Biomedical Engineering; Georgia Institute of Technology; Atlanta GA USA
- Department of Biomedical Engineering; Virginia Commonwealth University; Richmond VA USA
| | - Zvi Schwartz
- Wallace H. Coulter Department of Biomedical Engineering; Georgia Institute of Technology; Atlanta GA USA
- Department of Biomedical Engineering; Virginia Commonwealth University; Richmond VA USA
| |
Collapse
|
26
|
Sun D, Bu L, Liu C, Yin Z, Zhou X, Li X, Xiao A. Therapeutic effects of human amniotic fluid-derived stem cells on renal interstitial fibrosis in a murine model of unilateral ureteral obstruction. PLoS One 2013; 8:e65042. [PMID: 23724119 PMCID: PMC3665750 DOI: 10.1371/journal.pone.0065042] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 04/25/2013] [Indexed: 11/18/2022] Open
Abstract
Interstitial fibrosis is regarded as the main pathway for the progression of chronic kidney disease (CKD) and is often associated with severe renal dysfunction. Stem cell-based therapies may provide alternative approaches for the treatment of CKD. Human amniotic fluid-derived stem cells (hAFSCs) are a novel stem cell population, which exhibit both embryonic and mesenchymal stem cell characteristics. Herein, the present study investigated whether the transplantation of hAFSCs into renal tissues could improve renal interstitial fibrosis in a murine model of unilateral ureteral obstruction (UUO). We showed that hAFSCs provided a protective effect and alleviated interstitial fibrosis as reflected by an increase in microvascular density; additionally, hAFSCs treatment beneficially modulated protein levels of vascular endothelial growth factor (VEGF), hypoxia inducible factor-1α (HIF-1α) and transforming growth factor-β1 (TGF-β1). Therefore, we hypothesize that hAFSCs could represent an alternative, readily available source of stem cells that can be applied for the treatment of renal interstitial fibrosis.
Collapse
Affiliation(s)
- Dong Sun
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical College, Xuzhou, PR China.
| | | | | | | | | | | | | |
Collapse
|
27
|
Zavatti M, Resca E, Bertoni L, Maraldi T, Guida M, Carnevale G, Ferrari A, De Pol A. Ferutinin promotes proliferation and osteoblastic differentiation in human amniotic fluid and dental pulp stem cells. Life Sci 2013; 92:993-1003. [PMID: 23583571 DOI: 10.1016/j.lfs.2013.03.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 03/16/2013] [Accepted: 03/27/2013] [Indexed: 02/06/2023]
Abstract
AIMS The phytoestrogen Ferutinin plays an important role in prevention of osteoporosis caused by ovariectomy-induced estrogen deficiency in rats, but there is no evidence of its effect on osteoblastic differentiation in vitro. In this study we investigated the effect of Ferutinin on proliferation and osteoblastic differentiation of two different human stem cells populations, one derived from the amniotic fluid (AFSCs) and the other from the dental pulp (DPSCs). MAIN METHODS AFSCs and DPSCs were cultured in a differentiation medium for 14 or 21days with or without the addition of Ferutinin at a concentration ranging from 10(-11) to 10(-4)M. 17β-Estradiol was used as a positive drug at 10(-8)M. Cell proliferation and expression of specific osteoblast phenotype markers were analyzed. KEY FINDINGS MTT assay revealed that Ferutinin, at concentrations of 10(-8) and 10(-9)M, enhanced proliferation of both AFSCs and DPSCs after 72h of exposure. Moreover, in both stem cell populations, Ferutinin treatment induced greater expression of the osteoblast phenotype markers osteocalcin (OCN), osteopontin (OPN), collagen I, RUNX-2 and osterix (OSX), increased calcium deposition and osteocalcin secretion in the culture medium compared to controls. These effects were more pronounced after 14days of culture in both populations. SIGNIFICANCE The enhancing capabilities on proliferation and osteoblastic differentiation displayed by the phytoestrogen Ferutinin make this compound an interesting candidate to promote bone formation in vivo.
Collapse
Affiliation(s)
- M Zavatti
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplants, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
De Vita B, Campos LL, Listoni AJ, Maia L, Sudano MJ, Curcio BR, Landim-Alvarenga FC, Prestes NC. Isolamento, caracterização e diferenciação de células-tronco mesenquimais do líquido amniótico equino obtido em diferentes idades gestacionais. PESQUISA VETERINARIA BRASILEIRA 2013. [DOI: 10.1590/s0100-736x2013000400019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
O interesse nas pesquisas com células-tronco derivadas de anexos fetais de diversas espécies cresceu exponencialmente nas últimas décadas em virtude de serem fontes de células-tronco adultas com potencial de diferenciação em diversas linhagens celulares que apresentam pouca ou nenhuma imunogenicidade, apresentando-se assim como alternativa de grande importância para a formação de bancos celulares. Apesar do crescente interesse, os estudos para espécie equina ainda são escassos. O objetivo deste trabalho foi isolar, caracterizar e diferenciar células-tronco mesenquimais (CTMs) derivadas do líquido amniótico equino obtidas do terço inicial, médio e final da gestação (LA-CTMs), comparando suas características. Foram colhidas 23 amostras de líquido amniótico as quais foram submetidas às análises morfológica, imunocitoquímica, imunofenotípica por citometria de fluxo e às diferenciações osteogênica, adipogênica e condrogênica in vitro. Todas as amostras demonstraram adesão ao plástico e morfologia fibroblastóide. No ensaio imunocitoquímico as células de todos os grupos foram imunomarcadas para CD44, PCNA e vimentina com ausência de marcação para citoqueratina e Oct-4. Na citometria de fluxo observou-se a expressão de CD44 e CD90 e ausência de expressão de CD34, sendo que os marcadores CD44 e CD90 mostraram padrão de expressão decrescente em relação ao desenvolvimento gestacional. As amostras obtidas de todas as fases da gestação foram capazes de diferenciação nas linhagens osteogênica, condrogênica e adipogênica. Portanto, as células obtidas do líquido amniótico apresentaram características morfológicas, imunofenotípicas e potencial de diferenciação típicos das CTMs, demonstrando que a colheita pode ser realizada em qualquer fase gestacional. No entanto, mais pesquisas devem ser realizadas principalmente quanto à expressão de marcadores de pluripotencialidade (como o Oct-4) e ao seu potencial de diferenciação em linhagens extra mesodermais já relatados na literatura.
Collapse
|
29
|
Petsche Connell J, Camci-Unal G, Khademhosseini A, Jacot JG. Amniotic fluid-derived stem cells for cardiovascular tissue engineering applications. TISSUE ENGINEERING PART B-REVIEWS 2013; 19:368-79. [PMID: 23350771 DOI: 10.1089/ten.teb.2012.0561] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recent research has demonstrated that a population of stem cells can be isolated from amniotic fluid removed by amniocentesis that are broadly multipotent and nontumorogenic. These amniotic fluid-derived stem cells (AFSC) could potentially provide an autologous cell source for treatment of congenital defects identified during gestation, particularly cardiovascular defects. In this review, the various methods of isolating, sorting, and culturing AFSC are compared, along with techniques for inducing differentiation into cardiac myocytes and endothelial cells. Although research has not demonstrated complete and high-yield cardiac differentiation, AFSC have been shown to effectively differentiate into endothelial cells and can effectively support cardiac tissue. Additionally, several tissue engineering and regenerative therapeutic approaches for the use of these cells in heart patches, injection after myocardial infarction, heart valves, vascularized scaffolds, and blood vessels are summarized. These applications show great promise in the treatment of congenital cardiovascular defects, and further studies of isolation, culture, and differentiation of AFSC will help to develop their use for tissue engineering, regenerative medicine, and cardiovascular therapies.
Collapse
|
30
|
Kim J, Jeong SY, Ju YM, Yoo JJ, Smith TL, Khang G, Lee SJ, Atala A. In vitro osteogenic differentiation of human amniotic fluid-derived stem cells on a poly(lactide-co-glycolide) (PLGA)-bladder submucosa matrix (BSM) composite scaffold for bone tissue engineering. Biomed Mater 2013; 8:014107. [PMID: 23353783 DOI: 10.1088/1748-6041/8/1/014107] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Stem cells have become an important component of tissue regeneration, as they are able to differentiate into various cell types if guided appropriately. It is well known that cellular differentiation is greatly influenced by the surrounding microenvironment. We have developed a composite scaffold system using a collagen matrix derived from porcine bladder submucosa matrix (BSM) and poly(lactide-co-glycolide) (PLGA). In this study, we investigated whether a composite scaffold composed of naturally derived matrix combined with synthetic polymers would provide a microenvironment to facilitate the induction of osteogenic differentiation. We first showed that human amniotic fluid-derived stem cells (hAFSCs) adhered to the composite scaffolds and proliferated over time. We also showed that the composite scaffolds facilitated the differentiation of hAFSCs into an osteogenic lineage. The expression of osteogenic genes, including RUNX2, osteopontin (OPN) and osteocalcin (OCN) was upregulated in cells cultured on the composite scaffolds incubated in the osteogenic medium compared with ones without. Increased alkaline phosphatase (ALP) activity and calcium content indicates that hAFSCs seeded on 3D porous BSM-PLGA composite scaffolds resulted in higher mineralization rates as the duration of induction increased. This was also evidenced by the mineralized matrix within the scaffolds. The composite scaffold system provides a proper microenvironment that can facilitate osteogenic differentiation of AFSCs. This scaffold system may be a good candidate material for bone tissue engineering.
Collapse
Affiliation(s)
- Jaehyun Kim
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
de Lara Janz F, Favero GM, Bohatch MS, Aguiar Debes A, Bydlowski SP. Simvastatin induces osteogenic differentiation in human amniotic fluid mesenchymal stem cells (AFMSC). Fundam Clin Pharmacol 2012; 28:211-6. [PMID: 23094676 DOI: 10.1111/fcp.12006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 08/07/2012] [Accepted: 09/21/2012] [Indexed: 12/22/2022]
Abstract
Amniotic fluid is a complex mixture composed of water, salts and different cells types derived from embryo exfoliation. Some of these cells present similar characteristics to mesenchymal stem cells as adherent properties, typical surface antigens and differentiation capacity. These cells are called amniotic fluid-derived mesenchymal stem cells (AFMSCs) and are easily obtained by amniocentesis, propagated in culture and differentiated in several cell types with specific inductions. In this study, we observe the ability of simvastatin, a 3-HMG-CoA reductase inhibitor, to induce AFSMCs osteogenic differentiation. When AFSMCs were incubated with medium containing simvastatin, it was observed morphological changes, calcium deposits formation confirmed by Alizarin Red stain. Differentiated cells also expressed typical osteogenic genes, as osteopontin and osteocalcin. In conclusion, simvastatin could be used as an optional osteogenic induction agent for amniotic fluid-derived mesenchymal stem cells.
Collapse
Affiliation(s)
- Felipe de Lara Janz
- Laboratório de Genética e Hematologia Molecular (LIM 31), Faculdade de Medicina da, Universidade de São Paulo, São Paulo, SP, Brazil; Universidade Bandeirante de São Paulo - Anhanguera Educacional, São Bernardo do Campo, SP, Brazil
| | | | | | | | | |
Collapse
|
32
|
Rennie K, Gruslin A, Hengstschläger M, Pei D, Cai J, Nikaido T, Bani-Yaghoub M. Applications of amniotic membrane and fluid in stem cell biology and regenerative medicine. Stem Cells Int 2012; 2012:721538. [PMID: 23093978 PMCID: PMC3474290 DOI: 10.1155/2012/721538] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 09/07/2012] [Indexed: 12/16/2022] Open
Abstract
The amniotic membrane (AM) and amniotic fluid (AF) have a long history of use in surgical and prenatal diagnostic applications, respectively. In addition, the discovery of cell populations in AM and AF which are widely accessible, nontumorigenic and capable of differentiating into a variety of cell types has stimulated a flurry of research aimed at characterizing the cells and evaluating their potential utility in regenerative medicine. While a major focus of research has been the use of amniotic membrane and fluid in tissue engineering and cell replacement, AM- and AF-derived cells may also have capabilities in protecting and stimulating the repair of injured tissues via paracrine actions, and acting as vectors for biodelivery of exogenous factors to treat injury and diseases. Much progress has been made since the discovery of AM and AF cells with stem cell characteristics nearly a decade ago, but there remain a number of problematic issues stemming from the inherent heterogeneity of these cells as well as inconsistencies in isolation and culturing methods which must be addressed to advance the field towards the development of cell-based therapies. Here, we provide an overview of the recent progress and future perspectives in the use of AM- and AF-derived cells for therapeutic applications.
Collapse
Affiliation(s)
- Kerry Rennie
- Neurogenesis and Brain Repair, National Research Council-Institute for Biological Sciences, Bldg. M-54, Ottawa, ON, Canada K1A 0R6
| | - Andrée Gruslin
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada KIH 845
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada KIH 845
| | - Markus Hengstschläger
- Institute of Medical Genetics, Medical University of Vienna, Währinger Straße 10, 1090, Vienna, Austria
| | - Duanqing Pei
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou 510530, China
| | - Jinglei Cai
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou 510530, China
| | - Toshio Nikaido
- Department of Regenerative Medicine, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, 2630 Sugitani, Toyama 930-0194, Japan
| | - Mahmud Bani-Yaghoub
- Neurogenesis and Brain Repair, National Research Council-Institute for Biological Sciences, Bldg. M-54, Ottawa, ON, Canada K1A 0R6
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada KIH 845
| |
Collapse
|
33
|
Cananzi M, De Coppi P. CD117(+) amniotic fluid stem cells: state of the art and future perspectives. Organogenesis 2012; 8:77-88. [PMID: 23037870 DOI: 10.4161/org.22426] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Broadly multipotent stem cells can be isolated from amniotic fluid by selection for the expression of the membrane stem cell factor receptor c-Kit, a common marker for multipotential stem cells. They have clonogenic capability and can be directed into a wide range of cell types representing the three primary embryonic lineages. Amniotic fluid stem cells maintained for over 250 population doublings retained long telomeres and a normal karyotype. Clonal human lines verified by retroviral marking were induced to differentiate into cell types representing each embryonic germ layer, including cells of adipogenic, osteogenic, myogenic, endothelial, neuronal and hepatic lineages. AFS cells could be differentiate toward cardiomyogenic lineages, when co-cultured with neonatal cardiomyocytes, and have the potential to generate myogenic and hematopoietic lineages both in vitro and in vivo. Very recently first trimester AFS cells could be reprogrammed without any genetic manipulation opening new possibilities in the field of fetal/neonatal therapy and disease modeling. In this review we are aiming to summarize the knowledge on amniotic fluid stem cells and highlight the most promising results.
Collapse
Affiliation(s)
- Mara Cananzi
- Department of Paediatric Surgery, UCL Institute of Child Health & Great Ormond Street Hospital, London, UK
| | | |
Collapse
|
34
|
Joo S, Ko IK, Atala A, Yoo JJ, Lee SJ. Amniotic fluid-derived stem cells in regenerative medicine research. Arch Pharm Res 2012; 35:271-80. [PMID: 22370781 DOI: 10.1007/s12272-012-0207-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 12/07/2011] [Accepted: 12/07/2011] [Indexed: 12/21/2022]
Abstract
The stem cells isolated from amniotic fluid present an exciting possible contribution to the field of regenerative medicine and amniotic fluid-derived stem (AFS) cells have significant potential for research and therapeutic applications. AFS cells are multipotent, showing the ability to differentiate into cell types from all three embryonic germ layers. They express both embryonic and adult stem cell markers, expand extensively without feeder cells, double in 36 h, and are not tumorigenic. The AFS cells can be maintained for over 250 population doublings and preserve their telomere length and a normal karyotype. They differentiate easily into specific cell lineages and do not require human embryo tissue for their isolation, thus avoiding the current controversies associated with the use of human embryonic stem (ES) cells. The discovery of the AFS cells has been recent, and a great deal of work remains to be performed on the characterization and use of these cells. This review describes the various differentiated lineages that AFS cells can form and the future of these promising new stem cells in regenerative medicine research.
Collapse
Affiliation(s)
- Sunyoung Joo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157, USA
| | | | | | | | | |
Collapse
|
35
|
Baghaban Eslaminejad M, Jahangir S. Amniotic fluid stem cells and their application in cell-based tissue regeneration. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2012; 6:147-56. [PMID: 24520432 PMCID: PMC3850304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/08/2012] [Accepted: 05/07/2012] [Indexed: 11/20/2022]
Abstract
Advances in stem cell biotechnology hold great promise in the field of tissue engineering and regenerative medicine. Of interest are marrow mesenchymal stem cells (MSCs), embryonic stem cells (ESCs), and induced pluripotent stem cells (iPSCs). In addition, amniotic fluid stem cells (AFSCs) have attracted attention as a viable choice following the search for an alternative stem cell source. Investigators are interested in these cells because they come from the amniotic fluid that is routinely discarded after birth. There have been multiple investigations conducted worldwide in an attempt to better understand AF-SCs in terms of their potential use in regenerative medicine. In this review we give a brief introduction of amniotic fluid followed by a description of the cells present within this fluid. Their history related to stem cell discovery in the amniotic fluid as well as the main characteristics of AF-SCs are discussed. Finally, we elaborate on the potential for these cells to promote regeneration of various tissue defects, including fetal tissue, the nervous system, heart, lungs, kidneys, bones, and cartilage.
Collapse
Affiliation(s)
- Mohamadreza Baghaban Eslaminejad
- * Corresponding Address:
P.O. Box: 16635-148Department of Stem
Cells and Developmental BiologyCell Science Research CenterRoyan
Institute for Stem Cell Biology and TechnologyACECRTehranIran
| | | |
Collapse
|
36
|
Maraldi T, Riccio M, Resca E, Pisciotta A, La Sala GB, Ferrari A, Bruzzesi G, Motta A, Migliaresi C, Marzona L, De Pol A. Human amniotic fluid stem cells seeded in fibroin scaffold produce in vivo mineralized matrix. Tissue Eng Part A 2011; 17:2833-43. [PMID: 21864161 DOI: 10.1089/ten.tea.2011.0062] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
This study investigated the potential of amniotic fluid stem cells (AFSCs) to synthesize mineralized extracellular matrix (ECM) within different porous scaffolds of collagen, poly-D,L-lactic acid (PDLLA), and silk fibroin. The AFSCs were initially differentiated by using an osteogenic medium in two-dimensional culture, and expression of specific bone proteins and the physiologic mineral production by the AFSCs were analyzed. In particular, during differentiation process, AFSCs expressed proteins like Runt-related transcription factor 2 (Runx2), Osterix, Osteopontin, and Osteocalcin with a sequential expression, analogous to those occurring during osteoblast differentiation, and produced extracellular calcium stores. AFSCs were then cultured on three-dimensional (3D) scaffolds and evaluated for their ability to differentiate into osteoblastic cells in vivo. Stem cells were cultured in vitro for 1 week in collagen, fibroin, and PDLLA scaffolds. The effect of predifferentiation of the stem cells in scaffolds on the subsequent bone formation in vivo was determined in a rat subcutaneous model. With the addition of a third dimension, osteogenic differentiation and mineralized ECM production by AFSCs were significantly higher. This study demonstrated the strong potential of AFSCs to produce 3D mineralized bioengineered constructs in vivo and suggests that fibroin may be an effective scaffold material for functional repair of critical size bone defects.
Collapse
Affiliation(s)
- Tullia Maraldi
- Department of Laboratories, Pathological Anatomy and Forensic Medicine, University of Modena and Reggio Emilia, Modena, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Functional restoration of critically sized segmental defects with bone morphogenetic protein-2 and heparin treatment. Clin Orthop Relat Res 2011; 469:3111-7. [PMID: 21863396 PMCID: PMC3183200 DOI: 10.1007/s11999-011-2012-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Bone defects and fracture nonunions remain a substantial challenge for clinicians. Grafting procedures are limited by insufficient volume and donor site morbidity. As an alternative, biomaterial scaffolds functionalized through incorporation of growth factors such as bone morphogenetic proteins (BMPs) have been developed and appear to regenerate the structure and function of damaged or degenerated skeletal tissue. OBJECTIVES/PURPOSES Our objectives were therefore to determine whether: (1) the addition of heparin alone to collagen scaffolds sufficed to promote bone formation in vivo; (2) collagen-heparin scaffold improved BMP-mediated bone regeneration; and (3) precomplexed heparin and BMP-2 delivered on collagen scaffold could restore long bone biomechanical strength. METHODS We created bilateral surgical defects in the femora of 20 rats and filled the defects with PCL scaffolds with one of five treatments: collagen matrix (n = 5), collagen/heparin matrix (n = 7), collagen matrix + BMP-2 (n = 9), collagen/heparin matrix + BMP-2 (n = 9), or collagen matrix + BMP-2/heparin complex (n = 9). Bone formation was observed with radiographs and micro-CT analysis and biomechanical testing was used to assess strength. RESULTS The addition of heparin alone to collagen did not promote bone ingrowth and the addition of heparin to collagen did not improve BMP-mediated bone regeneration. Delivery of precomplexed BMP-2 and heparin in a collagen matrix resulted in new bone formation with mechanical properties similar to those of intact bone. CLINICAL RELEVANCE Our findings suggest delivery of precomplexed BMP-2 and heparin may be an advantageous strategy for treatment of clinically challenging bone defects.
Collapse
|
38
|
Dosier CR, Erdman CP, Park JH, Schwartz Z, Boyan BD, Guldberg RE. Resveratrol effect on osteogenic differentiation of rat and human adipose derived stem cells in a 3-D culture environment. J Mech Behav Biomed Mater 2011; 11:112-22. [PMID: 22658160 DOI: 10.1016/j.jmbbm.2011.08.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2011] [Revised: 08/15/2011] [Accepted: 08/19/2011] [Indexed: 12/25/2022]
Abstract
The goal of this study was to investigate the effect of resveratrol treatment on the osteogenic potential of human and rat adipose derived stem cells in a 3-D culture environment. Adipose derived stem cells (ADSCs) have been widely studied and have shown promise as a potential source of osteogenic progenitor cells. Previous work had investigated the effect of 25 μM resveratrol on the osteogenic differentiation of rat ADSCs in a 3-D environment and found that pre-treating cells for one passage prior to seeding on the scaffold yielded significantly more mineralization than untreated cells. We first sought to investigate whether this result was also observable with human ADSCs and found that the human cells did not respond to 25 μM resveratrol in a positive manner suggesting a species specific difference in resveratrol dosage. Therefore, we next investigated multiple doses at or below 25 μM resveratrol for both rat and human ADSCs. We found that doses below 25 μM caused significantly more mineralization than 0 (untreated) and 25 μM treated cells in a 3-D culture environment. Further, we observed species differences in the total amount of mineralized matrix, as well as the mean mineral density suggesting that the nature of mineralization of the extracellular matrix was different between species. Histological examination of the scaffolds showed that the human cell constructs remain highly cellular in nature with small pockets of mineralization, while rat cell constructs showed much larger and more mature mineralized nodules. Taken together, we demonstrate dose dependent differences in the mineralization response of human and rat ADSCs to resveratrol treatment, suggesting that in vitro pre-conditioning of 3D adipose-derived stem cell constructs may be an effective strategy to promote osteogenic differentiation prior to implantation.
Collapse
Affiliation(s)
- Christopher R Dosier
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
| | | | | | | | | | | |
Collapse
|
39
|
Park JS, Shim MS, Shim SH, Yang HN, Jeon SY, Woo DG, Lee DR, Yoon TK, Park KH. Chondrogenic potential of stem cells derived from amniotic fluid, adipose tissue, or bone marrow encapsulated in fibrin gels containing TGF-β3. Biomaterials 2011; 32:8139-49. [PMID: 21840589 DOI: 10.1016/j.biomaterials.2011.07.043] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Accepted: 07/13/2011] [Indexed: 12/16/2022]
Abstract
In this study, several types of hMSCs, derived from bone marrow, adipose tissue, or amniotic fluid, were encapsulated in a fibrin hydrogel mixed with TGF-β3 and then evaluated for their capacity for differentiation in vitro and in vivo. For determination of stem cell differentiation, RT-PCR, real time quantitative PCR (qPCR), histology, and immunohistochemical assays were used for analysis of chondrogenesis. Using these analysis methods, several of the cultured hMSCS were found to highly express genes and proteins specific to cartilage forming tissues. Additionally, similar trends in expression were found in tissue recovered from nude mice transplanted with several types of hMSCs encapsulated in a fibrin hydrogel containing TGF-β3. The results of both in vitro and in vivo analyses showed that cultured or transplanted hMSCs mixed with TGF-β3 in a fibrin hydrogel differentiated into chondrocytes, suggesting that these cells would be suitable for reconstruction of hyaline articular cartilage.
Collapse
Affiliation(s)
- Ji Sun Park
- Department of Biomedical Science, College of Life Science, CHA University 606-16, Yeoksam 1-dong, Gangnam-gu, Seoul 135-081, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Badylak SF, Taylor D, Uygun K. Whole-organ tissue engineering: decellularization and recellularization of three-dimensional matrix scaffolds. Annu Rev Biomed Eng 2011; 13:27-53. [PMID: 21417722 PMCID: PMC10887492 DOI: 10.1146/annurev-bioeng-071910-124743] [Citation(s) in RCA: 705] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The definitive treatment for end-stage organ failure is orthotopic transplantation. However, the demand for transplantation far exceeds the number of available donor organs. A promising tissue-engineering/regenerative-medicine approach for functional organ replacement has emerged in recent years. Decellularization of donor organs such as heart, liver, and lung can provide an acellular, naturally occurring three-dimensional biologic scaffold material that can then be seeded with selected cell populations. Preliminary studies in animal models have provided encouraging results for the proof of concept. However, significant challenges for three-dimensional organ engineering approach remain. This manuscript describes the fundamental concepts of whole-organ engineering, including characterization of the extracellular matrix as a scaffold, methods for decellularization of vascular organs, potential cells to reseed such a scaffold, techniques for the recellularization process and important aspects regarding bioreactor design to support this approach. Critical challenges and future directions are also discussed.
Collapse
Affiliation(s)
- Stephen F Badylak
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | | | | |
Collapse
|
41
|
Robey PG. Cell sources for bone regeneration: the good, the bad, and the ugly (but promising). TISSUE ENGINEERING PART B-REVIEWS 2011; 17:423-30. [PMID: 21797663 DOI: 10.1089/ten.teb.2011.0199] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Based on the extensive investigation of various ways to regenerate bone, bone marrow stromal cells, in conjunction with ceramic scaffolds, show great promise for application in human patients, and are already in use in a limited number of clinical trials. In preparing for clinical trials, scale-up current good manufacturing processes (cGMP) must incorporate the use of appropriate assays to ensure that the resulting cell product has maintained its biological activity. Future developments are needed to identify better scaffolds, and better ways to deliver cells with either injectable carriers, or by developing techniques to aide in their escape from the circulation and their incorporation into the pre-existing tissue. Lastly, development of methods that faithfully direct pluripotent stem cell differentiation into populations of osteogenic precursors (and ideally, containing skeletal stem cells) represents a new challenge in the field of bone regeneration, but also offer new opportunities to not only to study the biology of bone formation, but also to develop a robust cell source for bone regeneration.
Collapse
Affiliation(s)
- Pamela Gehron Robey
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland 20892, USA.
| |
Collapse
|
42
|
Dupont KM, Boerckel JD, Stevens HY, Diab T, Kolambkar YM, Takahata M, Schwarz EM, Guldberg RE. Synthetic scaffold coating with adeno-associated virus encoding BMP2 to promote endogenous bone repair. Cell Tissue Res 2011; 347:575-88. [PMID: 21695398 DOI: 10.1007/s00441-011-1197-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Accepted: 05/17/2011] [Indexed: 01/29/2023]
Abstract
Biomaterial scaffolds functionalized to stimulate endogenous repair mechanisms via the incorporation of osteogenic cues offer a potential alternative to bone grafting for the treatment of large bone defects. We first quantified the ability of a self-complementary adeno-associated viral vector encoding bone morphogenetic protein 2 (scAAV2.5-BMP2) to enhance human stem cell osteogenic differentiation in vitro. In two-dimensional culture, scAAV2.5-BMP2-transduced human mesenchymal stem cells (hMSCs) displayed significant increases in BMP2 production and alkaline phosphatase activity compared with controls. hMSCs and human amniotic-fluid-derived stem cells (hAFS cells) seeded on scAAV2.5-BMP2-coated three-dimensional porous polymer Poly(ε-caprolactone) (PCL) scaffolds also displayed significant increases in BMP2 production compared with controls during 12 weeks of culture, although only hMSC-seeded scaffolds displayed significantly increased mineral formation. PCL scaffolds coated with scAAV2.5-BMP2 were implanted into critically sized immunocompromised rat femoral defects, both with or without pre-seeding of hMSCs, representing ex vivo and in vivo gene therapy treatments, respectively. After 12 weeks, defects treated with acellular scAAV2.5-BMP2-coated scaffolds displayed increased bony bridging and had significantly higher bone ingrowth and mechanical properties compared with controls, whereas defects treated with scAAV2.5-BMP2 scaffolds pre-seeded with hMSCs failed to display significant differences relative to controls. When pooled, defect treatment with scAAV2.5-BMP2-coated scaffolds, both with or without inclusion of pre-seeded hMSCs, led to significant increases in defect mineral formation at all time points and increased mechanical properties compared with controls. This study thus presents a novel acellular bone-graft-free endogenous repair therapy for orthotopic tissue-engineered bone regeneration.
Collapse
Affiliation(s)
- Kenneth M Dupont
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30332, USA.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Klemmt PAB, Vafaizadeh V, Groner B. The potential of amniotic fluid stem cells for cellular therapy and tissue engineering. Expert Opin Biol Ther 2011; 11:1297-314. [DOI: 10.1517/14712598.2011.587800] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
44
|
Peister A, Woodruff MA, Prince JJ, Gray DP, Hutmacher DW, Guldberg RE. Cell sourcing for bone tissue engineering: amniotic fluid stem cells have a delayed, robust differentiation compared to mesenchymal stem cells. Stem Cell Res 2011; 7:17-27. [PMID: 21531647 DOI: 10.1016/j.scr.2011.03.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 03/08/2011] [Accepted: 03/08/2011] [Indexed: 12/16/2022] Open
Abstract
Cell based therapies for bone regeneration are an exciting emerging technology, but the availability of osteogenic cells is limited and an ideal cell source has not been identified. Amniotic fluid-derived stem cells (AFS) and bone-marrow derived mesenchymal stem cells (MSCs) were compared to determine their osteogenic differentiation capacity in both 2D and 3D environments. In 2D culture, the AFS cells produced more mineralized matrix but delayed peaks in osteogenic markers. Cells were also cultured on 3D scaffolds constructed of poly-ε-caprolactone for 15 weeks. MSCs differentiated more quickly than AFS cells on 3D scaffolds, but mineralized matrix production slowed considerably after 5 weeks. In contrast, the rate of AFS cell mineralization continued to increase out to 15 weeks, at which time AFS constructs contained 5-fold more mineralized matrix than MSC constructs. Therefore, cell source should be taken into consideration when used for cell therapy, as the MSCs would be a good choice for immediate matrix production, but the AFS cells would continue robust mineralization for an extended period of time. This study demonstrates that stem cell source can dramatically influence the magnitude and rate of osteogenic differentiation in vitro.
Collapse
Affiliation(s)
- Alexandra Peister
- Department of Biology, Morehouse College, 830 Westview Dr. SW, Atlanta, GA 30314, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Arnhold S, Glüer S, Hartmann K, Raabe O, Addicks K, Wenisch S, Hoopmann M. Amniotic-Fluid Stem Cells: Growth Dynamics and Differentiation Potential after a CD-117-Based Selection Procedure. Stem Cells Int 2011; 2011:715341. [PMID: 21437196 PMCID: PMC3062157 DOI: 10.4061/2011/715341] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 10/25/2010] [Accepted: 01/09/2011] [Indexed: 12/16/2022] Open
Abstract
Amniotic fluid (AF) has become an interesting source of fetal stem cells. However, AF contains heterogeneous and multiple, partially differentiated cell types. After isolation from the amniotic fluid, cells were characterized regarding their morphology and growth dynamics. They were sorted by magnetic associated cell sorting using the surface marker CD 117. In order to show stem cell characteristics such as pluripotency and to evaluate a possible therapeutic application of these cells, AF fluid-derived stem cells were differentiated along the adipogenic, osteogenic, and chondrogenic as well as the neuronal lineage under hypoxic conditions. Our findings reveal that magnetic associated cell sorting (MACS) does not markedly influence growth characteristics as demonstrated by the generation doubling time. There was, however, an effect regarding an altered adipogenic, osteogenic, and chondrogenic differentiation capacity in the selected cell fraction. In contrast, in the unselected cell population neuronal differentiation is enhanced.
Collapse
Affiliation(s)
- S Arnhold
- Department of Veterinary Anatomy, Justus-Liebig-University Giessen, Frankfurter Straße 98, 35392 Giessen, Germany
| | | | | | | | | | | | | |
Collapse
|
46
|
Kolambkar YM, Peister A, Ekaputra AK, Hutmacher DW, Guldberg RE. Colonization and osteogenic differentiation of different stem cell sources on electrospun nanofiber meshes. Tissue Eng Part A 2011; 16:3219-30. [PMID: 20504075 DOI: 10.1089/ten.tea.2010.0004] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Numerous challenges remain in the successful clinical translation of cell-based therapies for musculoskeletal tissue repair, including the identification of an appropriate cell source and a viable cell delivery system. The aim of this study was to investigate the attachment, colonization, and osteogenic differentiation of two stem cell types, human mesenchymal stem cells (hMSCs) and human amniotic fluid stem (hAFS) cells, on electrospun nanofiber meshes. We demonstrate that nanofiber meshes are able to support these cell functions robustly, with both cell types demonstrating strong osteogenic potential. Differences in the kinetics of osteogenic differentiation were observed between hMSCs and hAFS cells, with the hAFS cells displaying a delayed alkaline phosphatase peak, but elevated mineral deposition, compared to hMSCs. We also compared the cell behavior on nanofiber meshes to that on tissue culture plastic, and observed that there is delayed initial attachment and proliferation on meshes, but enhanced mineralization at a later time point. Finally, cell-seeded nanofiber meshes were found to be effective in colonizing three-dimensional scaffolds in an in vitro system. This study provides support for the use of the nanofiber mesh as a model surface for cell culture in vitro, and a cell delivery vehicle for the repair of bone defects in vivo.
Collapse
Affiliation(s)
- Yash M Kolambkar
- Wallace H. Coulter Department of Biomedical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | | | | | | | | |
Collapse
|
47
|
Rai B, Lin JL, Lim ZX, Guldberg RE, Hutmacher DW, Cool SM. Differences between in vitro viability and differentiation and in vivo bone-forming efficacy of human mesenchymal stem cells cultured on PCL–TCP scaffolds. Biomaterials 2010; 31:7960-70. [DOI: 10.1016/j.biomaterials.2010.07.001] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Accepted: 07/01/2010] [Indexed: 01/13/2023]
|
48
|
|
49
|
Yeh YC, Wei HJ, Lee WY, Yu CL, Chang Y, Hsu LW, Chung MF, Tsai MS, Hwang SM, Sung HW. Cellular cardiomyoplasty with human amniotic fluid stem cells: in vitro and in vivo studies. Tissue Eng Part A 2010; 16:1925-36. [PMID: 20067384 DOI: 10.1089/ten.tea.2009.0728] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Human amniotic fluid stem cells (hAFSCs) derived from second-trimester amniocentesis were evaluated for the therapeutic potential of cardiac repair. Whether hAFSCs can be differentiated into cardiomyogenic cells and toward the maturation of endothelial cell lineage was investigated in vitro using mimicking differentiation milieu. Employing an immune-suppressed rat model with experimental myocardial infarction, an intramyocardial injection was conducted with a needle directly into the peri-infarct areas. There were three treatment groups: sham, saline, and hAFSCs (n > or = 10). When cultured with rat neonatal cardiomyocytes or in endothelial growth medium-2 enriched with vascular endothelial growth factor, hAFSCs were differentiated into cardiomyocyte-like cells and cells of endothelial lineage, respectively. After 4 weeks, hAFSC-treated animals showed a preservation of the infarcted thickness, an attenuation of left ventricle remodeling, a higher vascular density, and thus an improvement in cardiac function, when compared with the saline injection group. Survival and proliferation of the transplanted hAFSCs were revealed by immunohistochemical staining. Expressions of the cardiac-specific markers such as Nkx2.5, alpha-actinin, and cardiac Troponin T were observed in the transplanted hAFSCs. Additionally, Cx43 was clearly expressed at the borders of the transplanted/transplanted and host/transplanted cells, an indication of enhancement of cell connection. The results demonstrated that hAFSCs induce angiogenesis, have cardiomyogenic potential, and may be used as a new cell source for cellular cardiomyoplasty.
Collapse
Affiliation(s)
- Yi-Chun Yeh
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Human stem cell delivery for treatment of large segmental bone defects. Proc Natl Acad Sci U S A 2010; 107:3305-10. [PMID: 20133731 DOI: 10.1073/pnas.0905444107] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Local or systemic stem cell delivery has the potential to promote repair of a variety of damaged or degenerated tissues. Although various stem cell sources have been investigated for bone repair, few comparative reports exist, and cellular distribution and viability postimplantation remain key issues. In this study, we quantified the ability of tissue-engineered constructs containing either human fetal or adult stem cells to enhance functional repair of nude rat critically sized femoral defects. After 12 weeks, defects treated with cell-seeded polymer scaffolds had significantly higher bone ingrowth and torsional strength compared to those receiving acellular scaffolds, although there were no significant differences between the cell sources. Next, stem cells were labeled with fluorescent quantum dots (QDs) in an attempt to noninvasively track their distribution after delivery on scaffolds. Clear fluorescence was observed at implantation sites throughout the study; however, beginning 7-10 days after surgery, signals were also observed at contralateral sites treated with acellular QD-free scaffolds. Although immunostaining for human nuclei revealed retention of some cells at the implantation site, no human cells were detected in the control limb defects. Additional histological analysis of implantation and control defect tissues revealed macrophages containing endocytosed QDs. Furthermore, QD-labeling appeared to diminish transplanted cell function resulting in reduced healing responses. In summary, augmentation of polymeric scaffolds with stem cells derived from fetal and adult tissues significantly enhanced healing of large segmental bone defects; however, QD labeling of stem cells eliminated the observed therapeutic effect and failed to conclusively track stem cell location long-term in vivo.
Collapse
|