1
|
Ramzan F, Salim A, Hussain A, Khan I. Unleashing the Healing Power of Mesenchymal Stem Cells for Osteochondral Abnormalities. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2024. [DOI: 10.1007/s40883-024-00356-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 07/25/2024] [Accepted: 08/31/2024] [Indexed: 01/11/2025]
|
2
|
Ma L, Dong W, Lai E, Wang J. Silk fibroin-based scaffolds for tissue engineering. Front Bioeng Biotechnol 2024; 12:1381838. [PMID: 38737541 PMCID: PMC11084674 DOI: 10.3389/fbioe.2024.1381838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 04/12/2024] [Indexed: 05/14/2024] Open
Abstract
Silk fibroin is an important natural fibrous protein with excellent prospects for tissue engineering applications. With profound studies in recent years, its potential in tissue repair has been developed. A growing body of literature has investigated various fabricating methods of silk fibroin and their application in tissue repair. The purpose of this paper is to trace the latest developments of SF-based scaffolds for tissue engineering. In this review, we first presented the primary and secondary structures of silk fibroin. The processing methods of SF scaffolds were then summarized. Lastly, we examined the contribution of new studies applying SF as scaffolds in tissue regeneration applications. Overall, this review showed the latest progress in the fabrication and utilization of silk fibroin-based scaffolds.
Collapse
Affiliation(s)
- Li Ma
- National Innovation Center for Advanced Medical Devices, Shenzhen, China
| | - Wenyuan Dong
- National Innovation Center for Advanced Medical Devices, Shenzhen, China
| | - Enping Lai
- College of Biological and Chemical Engineering, Guangxi University of Science and Technology, Liuzhou, China
| | - Jiamian Wang
- National Innovation Center for Advanced Medical Devices, Shenzhen, China
| |
Collapse
|
3
|
Pereira L, Echarte L, Romero M, Grazioli G, Pérez-Campos H, Francia A, Vicentino W, Mombrú AW, Faccio R, Álvarez I, Touriño C, Pardo H. Synthesis and characterization of a bovine collagen: GAG scaffold with Uruguayan raw material for tissue engineering. Cell Tissue Bank 2024; 25:123-142. [PMID: 34536180 DOI: 10.1007/s10561-021-09960-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 09/06/2021] [Indexed: 11/28/2022]
Abstract
Tissue engineering (TE) and regenerative medicine offer strategies to improve damaged tissues by using scaffolds and cells. The use of collagen-based biomaterials in the field of TE has been intensively growing over the past decades. Mesenchymal stromal cells (MSCs) and dental pulp stem cells (DPSCs) are promising cell candidates for development of clinical composites. In this study, we proposed the development of a bovine collagen type I: chondroitin-6-sulphate (CG) scaffold, obtained from Uruguayan raw material (certified as free bovine spongiform encephalopathy), with CG crosslinking enhancement using different gamma radiation doses. Structural, biomechanical and chemical characteristics of the scaffolds were assessed by Scanning Electron Microscopy, axial tensile tests, FT-IR and Raman Spectroscopy, respectively. Once we selected the most appropriate scaffold for future use as a TE product, we studied the behavior of MSCs and DPSCs cultured on the scaffold by cytotoxicity, proliferation and differentiation assays. Among the diverse porous scaffolds obtained, the one with the most adequate properties was the one exposed to 15 kGy of gamma radiation. This radiation dose contributed to the crosslinking of molecules, to the formation of new bonds and/or to the reorganization of the collagen fibers. The selected scaffold was non-cytotoxic for the tested cells and a suitable substrate for cell proliferation. Furthermore, the scaffold allowed MSCs differentiation to osteogenic, chondrogenic, and adipogenic lineages. Thus, this work shows a promising approach to the synthesis of a collagen-scaffold suitable for TE.
Collapse
Affiliation(s)
- L Pereira
- Centro NanoMat, Facultad de Química, Instituto Polo Tecnológico de Pando, UdelaR, Camino Aparicio Saravia s/n, 9100, Pando, Canelones, Uruguay
| | - L Echarte
- Área Terapia Celular y Medicina Regenerativa (ATCMR), Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - M Romero
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República (UdelaR), General Flores, 2124, 11800, Montevideo, Uruguay
| | - G Grazioli
- Cátedra de Materiales Dentales, Facultad de Odontología, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - H Pérez-Campos
- Instituto Nacional de Donación y Trasplante (INDT), Ministerio de salud Pública-Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Ministerio, Uruguay
| | - A Francia
- Fisiología general y bucodental, Facultad de Odontología, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - W Vicentino
- Instituto Nacional de Donación y Trasplante (INDT), Ministerio de salud Pública-Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Ministerio, Uruguay
| | - A W Mombrú
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República (UdelaR), General Flores, 2124, 11800, Montevideo, Uruguay
| | - R Faccio
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República (UdelaR), General Flores, 2124, 11800, Montevideo, Uruguay
| | - I Álvarez
- Instituto Nacional de Donación y Trasplante (INDT), Ministerio de salud Pública-Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Ministerio, Uruguay
| | - C Touriño
- Área Terapia Celular y Medicina Regenerativa (ATCMR), Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Uruguay.
| | - H Pardo
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República (UdelaR), General Flores, 2124, 11800, Montevideo, Uruguay.
| |
Collapse
|
4
|
Pishnamazi SM, Ghaderian SMH, Irani S, Ardeshirylajimi A. Polycaprolactone/poly L-lactic acid nanofibrous scaffold improves osteogenic differentiation of the amniotic fluid-derived stem cells. In Vitro Cell Dev Biol Anim 2024; 60:106-114. [PMID: 38123755 DOI: 10.1007/s11626-023-00838-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023]
Abstract
Using stem cells is one of the most important determining factors in repairing lesions using regenerative medicine. Obtaining adult stem cells from patients is a perfect choice, but it is worth noting that their differentiation and proliferation potential decreases as the patient ages. For this reason, the use of amniotic fluid stem cells can be one of the excellent alternatives. This research aimed to investigate the osteogenic differentiation potential of the amniotic fluid stem cells while cultured on the polycaprolactone/poly L-lactic acid nanofibrous scaffold. Scaffolds were qualitatively evaluated by a scanning electron microscope, and their hydrophilicity and mechanical properties were studied using contact angle and tensile test, respectively. The biocompatibility and non-toxicity of the nanofibers were also evaluated using viability assay. The osteo-supportive capacity of the nanofibers was examined using alizarin red staining, alkaline phosphatase activity, and calcium release measurement. Finally, the expression level of four important bone-related genes was determined quantitatively. The results demonstrated that the mineralization rate, alkaline phosphatase activity, intracellular calcium, and bone-related genes increased significantly in the cells cultured on the polycaprolactone/poly L-lactic acid scaffold compared to the cells cultured on the tissue culture plate as a control. According to the results, it can be concluded that the polycaprolactone/poly L-lactic acid nanofibrous scaffold surprisingly improved the osteogenic differentiation potential of the amniotic fluid stem cells and, in combination with polycaprolactone/poly L-lactic acid nanofibers could be a promising candidate as bone implants.
Collapse
Affiliation(s)
| | | | - Shiva Irani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | |
Collapse
|
5
|
Fu JN, Wang X, Yang M, Chen YR, Zhang JY, Deng RH, Zhang ZN, Yu JK, Yuan FZ. Scaffold-Based Tissue Engineering Strategies for Osteochondral Repair. Front Bioeng Biotechnol 2022; 9:812383. [PMID: 35087809 PMCID: PMC8787149 DOI: 10.3389/fbioe.2021.812383] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/16/2021] [Indexed: 12/19/2022] Open
Abstract
Over centuries, several advances have been made in osteochondral (OC) tissue engineering to regenerate more biomimetic tissue. As an essential component of tissue engineering, scaffolds provide structural and functional support for cell growth and differentiation. Numerous scaffold types, such as porous, hydrogel, fibrous, microsphere, metal, composite and decellularized matrix, have been reported and evaluated for OC tissue regeneration in vitro and in vivo, with respective advantages and disadvantages. Unfortunately, due to the inherent complexity of organizational structure and the objective limitations of manufacturing technologies and biomaterials, we have not yet achieved stable and satisfactory effects of OC defects repair. In this review, we summarize the complicated gradients of natural OC tissue and then discuss various osteochondral tissue engineering strategies, focusing on scaffold design with abundant cell resources, material types, fabrication techniques and functional properties.
Collapse
Affiliation(s)
- Jiang-Nan Fu
- Department of Sports Medicine, Peking University Third Hospital, Beijing, China.,Institute of Sports Medicine of Peking University, Beijing, China
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Meng Yang
- Department of Sports Medicine, Peking University Third Hospital, Beijing, China.,Institute of Sports Medicine of Peking University, Beijing, China
| | - You-Rong Chen
- Department of Sports Medicine, Peking University Third Hospital, Beijing, China.,Institute of Sports Medicine of Peking University, Beijing, China
| | - Ji-Ying Zhang
- Department of Sports Medicine, Peking University Third Hospital, Beijing, China.,Institute of Sports Medicine of Peking University, Beijing, China
| | - Rong-Hui Deng
- Department of Sports Medicine, Peking University Third Hospital, Beijing, China.,Institute of Sports Medicine of Peking University, Beijing, China
| | - Zi-Ning Zhang
- Department of Sports Medicine, Peking University Third Hospital, Beijing, China.,Institute of Sports Medicine of Peking University, Beijing, China
| | - Jia-Kuo Yu
- Department of Sports Medicine, Peking University Third Hospital, Beijing, China.,Institute of Sports Medicine of Peking University, Beijing, China
| | - Fu-Zhen Yuan
- Department of Sports Medicine, Peking University Third Hospital, Beijing, China.,Institute of Sports Medicine of Peking University, Beijing, China
| |
Collapse
|
6
|
Castelli V, Antonucci I, d'Angelo M, Tessitore A, Zelli V, Benedetti E, Ferri C, Desideri G, Borlongan C, Stuppia L, Cimini A. Neuroprotective effects of human amniotic fluid stem cells-derived secretome in an ischemia/reperfusion model. Stem Cells Transl Med 2021; 10:251-266. [PMID: 33027557 PMCID: PMC7848376 DOI: 10.1002/sctm.20-0268] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 08/03/2020] [Accepted: 08/24/2020] [Indexed: 12/11/2022] Open
Abstract
Stem cells offer the basis for the promotion of robust new therapeutic approaches for a variety of human disorders. There are still many limitations to be overcome before clinical therapeutic application, including a better understanding of the mechanism by which stem cell therapies may lead to enhanced recovery. In vitro investigations are necessary to dissect the mechanisms involved and to support the potential development in stem cell-based therapies. In spite of growing interest in human amniotic fluid stem cells, not much is known about the characteristics of their secretome and regarding the potential neuroprotective mechanism in different pathologies, including stroke. To get more insight on amniotic fluid cells therapeutic potential, signal transduction pathways activated by human amniotic fluid stem cells (hAFSCs)-derived secretome in a stroke in vitro model (ischemia/reperfusion [I/R] model) were investigated by Western blot. Moreover, miRNA expression in the exosomal fraction of the conditioned medium was analyzed. hAFSCs-derived secretome was able to activate pro-survival and anti-apoptotic pathways. MicroRNA analysis in the exosomal component revealed a panel of 16 overexpressed miRNAs involved in the regulation of coherent signaling pathways. In particular, the pathways of relevance in ischemia/reperfusion, such as neurotrophin signaling, and those related to neuroprotection and neuronal cell death, were analyzed. The results obtained strongly point toward the neuroprotective effects of the hAFSCs-conditioned medium in the in vitro stroke model here analyzed. This can be achieved by the modulation and activation of pro-survival processes, at least in part, due to the activity of secreted miRNAs.
Collapse
Affiliation(s)
- Vanessa Castelli
- Department of Life, Health and Environmental SciencesUniversity of L'AquilaL'AquilaItaly
| | - Ivana Antonucci
- Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences“G. d'Annunzio” UniversityChieti‐PescaraItaly
- Center for Advanced Studies and Technology (CAST)‘G. d'Annunzio’ UniversityChieti‐PescaraItaly
| | - Michele d'Angelo
- Department of Life, Health and Environmental SciencesUniversity of L'AquilaL'AquilaItaly
| | - Alessandra Tessitore
- Department of Biotechnological and Applied Clinical Sciences (DISCAB)University of L'AquilaL'AquilaItaly
| | - Veronica Zelli
- Department of Biotechnological and Applied Clinical Sciences (DISCAB)University of L'AquilaL'AquilaItaly
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental SciencesUniversity of L'AquilaL'AquilaItaly
| | - Claudio Ferri
- Department of Life, Health and Environmental SciencesUniversity of L'AquilaL'AquilaItaly
| | | | - Cesar Borlongan
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain RepairUniversity of South Florida College of MedicineTampaFloridaUSA
| | - Liborio Stuppia
- Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences“G. d'Annunzio” UniversityChieti‐PescaraItaly
- Center for Advanced Studies and Technology (CAST)‘G. d'Annunzio’ UniversityChieti‐PescaraItaly
| | - Annamaria Cimini
- Department of Life, Health and Environmental SciencesUniversity of L'AquilaL'AquilaItaly
- Sbarro Institute for Cancer Research and Molecular Medicine and Centre for BiotechnologyTemple UniversityPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
7
|
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
|
8
|
Casciaro F, Beretti F, Zavatti M, McCubrey JA, Ratti S, Marmiroli S, Follo MY, Maraldi T. Nuclear Nox4 interaction with prelamin A is associated with nuclear redox control of stem cell aging. Aging (Albany NY) 2019; 10:2911-2934. [PMID: 30362963 PMCID: PMC6224265 DOI: 10.18632/aging.101599] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 10/13/2018] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cells have emerged as an important tool that can be used for tissue regeneration thanks to their easy preparation, differentiation potential and immunomodulatory activity. However, an extensive culture of stem cells in vitro prior to clinical use can lead to oxidative stress that can modulate different stem cells properties, such as self-renewal, proliferation, differentiation and senescence. The aim of this study was to investigate the aging process occurring during in vitro expansion of stem cells, obtained from amniotic fluids (AFSC) at similar gestational age. The analysis of 21 AFSC samples allowed to classify them in groups with different levels of stemness properties. In summary, the expression of pluripotency genes and the proliferation rate were inversely correlated with the content of reactive oxygen species (ROS), DNA damage signs and the onset premature aging markers, including accumulation of prelamin A, the lamin A immature form. Interestingly, a specific source of ROS, the NADPH oxidase isoform 4 (Nox4), can localize into PML nuclear bodies (PML-NB), where it associates to prelamin A. Besides, Nox4 post translational modification, involved in PML-NB localization, is linked to its degradation pathway, as it is also for prelamin A, thus possibly modulating the premature aging phenotype occurrence.
Collapse
Affiliation(s)
- Francesca Casciaro
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, 41124, Italy.,Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, 40126, Italy
| | - Francesca Beretti
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, 41124, Italy
| | - Manuela Zavatti
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, 41124, Italy
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA
| | - Stefano Ratti
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, 40126, Italy
| | - Sandra Marmiroli
- Cellular Signaling Unit, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy
| | - Matilde Y Follo
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, 40126, Italy
| | - Tullia Maraldi
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, 41124, Italy
| |
Collapse
|
9
|
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
|
10
|
Hydrogen sulfide-releasing silk fibroin scaffold for bone tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 102:471-482. [DOI: 10.1016/j.msec.2019.04.039] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 03/20/2019] [Accepted: 04/12/2019] [Indexed: 02/07/2023]
|
11
|
Senel U, Coskun OS, Tuysuz EC, Sahin M, Bayrak OF, Cakmak B, Tanriverdi HI, Kuskucu A. Smooth muscle cell differentiation from rabbit amniotic cells. Exp Mol Pathol 2018; 105:395-403. [PMID: 30414978 DOI: 10.1016/j.yexmp.2018.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/14/2018] [Accepted: 11/06/2018] [Indexed: 11/25/2022]
Abstract
Amniotic fluid (AF) is the liquid layer that provides mechanical support and allows movement of the fetus during embryogenesis. Mesenchymal stem cells (MSCs), which have differentiation capacity, are also found in AF-derived cells at a low ratio. Smooth muscle cells (SMCs) play an important role in organ function and are frequently used in tissue engineering. We examined the differentiation of AF-derived MSCs (AMSCs) into SMCs. AMSCs were sorted from cultured amniotic cells and differentiated into SMCs using differentiation agents, including platelet-derived growth factor BB (PDGF-BB) and tumor growth factor β (TGF-β). Characterization of differentiated SMCs was confirmed morphologically, molecularly (via quantitative polymerase chain reaction [qPCR] and immunocytochemistry [ICC]), and functionally (using a contractile assay and fluo-4 calcium signaling assay). Poly(lactide-co-glycolide) (PLGA) scaffolds were fabricated, and the attachment capacity of AMSCs was assessed via scanning electron microscopy. AMSCs were successfully differentiated into SMCs. Our results indicate that AMSCs change their morphology and exhibit increased expression of ACTA2 and MYH11, which was confirmed via qPCR and ICC. Furthermore, functional experiments revealed that differentiated SMCs had both contraction ability and increased Ca2 concentration in the cytoplasm. Finally, PLGA scaffolds were prepared and AMSCs were successfully planted onto the scaffolds. The AMSCs fully differentiated into functional SMCs, and the PLGA polymer is a suitable scaffold material for AMSCs. With further clinical trials, AF-derived MSC-based SMC engineering may become a highly efficient treatment option.
Collapse
Affiliation(s)
- Ufuk Senel
- Department of Pediatric Surgery, Faculty of Medicine, Gaziosmanpasa University, 60100 Tokat, Turkey
| | - Ozlem Silan Coskun
- Department of Medical Genetics, Yeditepe University Medical School, 34755 Istanbul, Turkey; Department of Biotechnology, Institute of Science, Yeditepe University, 34755 Istanbul, Turkey
| | - Emre Can Tuysuz
- Department of Medical Genetics, Yeditepe University Medical School, 34755 Istanbul, Turkey; Department of Biotechnology, Institute of Science, Yeditepe University, 34755 Istanbul, Turkey
| | - Mesut Sahin
- Department of Nanoscience and Nanoengineering, Institute of Science Ataturk University, 25240 Erzurum, Turkey
| | - Omer Faruk Bayrak
- Department of Medical Genetics, Yeditepe University Medical School, 34755 Istanbul, Turkey
| | - Bulent Cakmak
- Department of Obstetrics and Gynecology, Faculty of Medicine, Gaziosmanpasa University, 60100 Tokat, Turkey
| | - Halil Ibrahim Tanriverdi
- Department of Pediatric Surgery, Faculty of Medicine, Gaziosmanpasa University, 60100 Tokat, Turkey
| | - Aysegul Kuskucu
- Department of Medical Genetics, Yeditepe University Medical School, 34755 Istanbul, Turkey.
| |
Collapse
|
12
|
Raggio R, Bonani W, Callone E, Dirè S, Gambari L, Grassi F, Motta A. Silk Fibroin Porous Scaffolds Loaded with a Slow-Releasing Hydrogen Sulfide Agent (GYY4137) for Applications of Tissue Engineering. ACS Biomater Sci Eng 2018; 4:2956-2966. [DOI: 10.1021/acsbiomaterials.8b00212] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Rosasilvia Raggio
- Department of Industrial Engineering, University of Trento, Via Sommarive 9, 38123 Trento, Italy
- BIOtech Research Center and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Via delle Regole 101, 38123 Trento, Italy
| | - Walter Bonani
- Department of Industrial Engineering, University of Trento, Via Sommarive 9, 38123 Trento, Italy
- BIOtech Research Center and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Via delle Regole 101, 38123 Trento, Italy
| | - Emanuela Callone
- Department of Industrial Engineering, University of Trento, Via Sommarive 9, 38123 Trento, Italy
- “Klaus Muller” Magnetic Resonance Laboratory, Department of Industrial Engineering, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Sandra Dirè
- Department of Industrial Engineering, University of Trento, Via Sommarive 9, 38123 Trento, Italy
- “Klaus Muller” Magnetic Resonance Laboratory, Department of Industrial Engineering, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Laura Gambari
- RAMSES Laboratory, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Francesco Grassi
- RAMSES Laboratory, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Antonella Motta
- Department of Industrial Engineering, University of Trento, Via Sommarive 9, 38123 Trento, Italy
- BIOtech Research Center and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Via delle Regole 101, 38123 Trento, Italy
| |
Collapse
|
13
|
Beretti F, Zavatti M, Casciaro F, Comitini G, Franchi F, Barbieri V, La Sala GB, Maraldi T. Amniotic fluid stem cell exosomes: Therapeutic perspective. Biofactors 2018; 44:158-167. [PMID: 29341292 DOI: 10.1002/biof.1407] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 11/16/2017] [Accepted: 12/04/2017] [Indexed: 12/11/2022]
Abstract
It is widely accepted that the therapeutic potential of stem cells can be largely mediated by paracrine factors, also included into exosomes. Thus, stem cell-derived exosomes represent a major therapeutic option in regenerative medicine avoiding, if compared to stem cells graft, abnormal differentiation and tumor formation. Exosomes derived from mesenchymal stem cells (MSC) induce damaged tissue repair, and can also exert immunomodulatory effects on the differentiation, activation and function of different lymphocytes. Therefore, MSC exosomes can be considered as a potential treatment for inflammatory diseases and also an ideal candidate for allogeneic therapy due to their low immunogenicity. Amniotic fluid stem cells (AFSCs) are broadly multipotent, can be expanded in culture, and can be easily cryopreserved in cellular banks. In this study, morphology, phenotype, and protein content of exosomes released into amniotic fluid in vivo and from AFSC during in vitro culture (conditioned medium) were examined. We found that AFSC-derived exosomes present different molecules than amniotic fluid ones, some of them involved in immunomodulation, such transforming growth factor beta and hepatic growth factors. The immunomodulatory effect of AFSC's exosomes on peripheral blood mononuclear cells stimulated with phytohemagglutinin was compared to that of the supernatant produced by such conditioned media deprived of exosomes. We present evidence that the principal effect of AFSC conditioned media (without exosomes) is the induction of apoptosis in lymphocytes, whereas exposure to AFSC-derived exosomes decreases the lymphocyte's proliferation, supporting the hypothesis that the entire secretome of stem cells differently affects immune-response. © 2017 BioFactors, 44(2):158-167, 2018.
Collapse
Affiliation(s)
- Francesca Beretti
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Manuela Zavatti
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesca Casciaro
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giuseppina Comitini
- Unit of Obstetrics & Gynecology, IRCCS-ASMN of Reggio Emilia, Reggio Emilia, Italy
| | - Fabrizia Franchi
- Genetic Laboratory, IRCCS-ASMN of Reggio Emilia, Reggio Emilia, Italy
| | - Veronica Barbieri
- Genetic Laboratory, IRCCS-ASMN of Reggio Emilia, Reggio Emilia, Italy
| | - Giovanni B La Sala
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Obstetrics & Gynecology, IRCCS-ASMN of Reggio Emilia, Reggio Emilia, Italy
| | - Tullia Maraldi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
14
|
Bhattacharjee P, Kundu B, Naskar D, Kim HW, Maiti TK, Bhattacharya D, Kundu SC. Silk scaffolds in bone tissue engineering: An overview. Acta Biomater 2017; 63:1-17. [PMID: 28941652 DOI: 10.1016/j.actbio.2017.09.027] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 08/26/2017] [Accepted: 09/19/2017] [Indexed: 12/17/2022]
Abstract
Bone tissue plays multiple roles in our day-to-day functionality. The frequency of accidental bone damage and disorder is increasing worldwide. Moreover, as the world population continues to grow, the percentage of the elderly population continues to grow, which results in an increased number of bone degenerative diseases. This increased elderly population pushes the need for artificial bone implants that specifically employ biocompatible materials. A vast body of literature is available on the use of silk in bone tissue engineering. The current work presents an overview of this literature from materials and fabrication perspective. As silk is an easy-to-process biopolymer; this allows silk-based biomaterials to be molded into diverse forms and architectures, which further affects the degradability. This makes silk-based scaffolds suitable for treating a variety of bone reconstruction and regeneration objectives. Silk surfaces offer active sites that aid the mineralization and/or bonding of bioactive molecules that facilitate bone regeneration. Silk has also been blended with a variety of polymers and minerals to enhance its advantageous properties or introduce new ones. Several successful works, both in vitro and in vivo, have been reported using silk-based scaffolds to regenerate bone tissues or other parts of the skeletal system such as cartilage and ligament. A growing trend is observed toward the use of mineralized and nanofibrous scaffolds along with the development of technology that allows to control scaffold architecture, its biodegradability and the sustained releasing property of scaffolds. Further development of silk-based scaffolds for bone tissue engineering, taking them up to and beyond the stage of human trials, is hoped to be achieved in the near future through a cross-disciplinary coalition of tissue engineers, material scientists and manufacturing engineers. STATEMENT OF SIGNIFICANCE The state-of-art of silk biomaterials in bone tissue engineering, covering their wide applications as cell scaffolding matrices to micro-nano carriers for delivering bone growth factors and therapeutic molecules to diseased or damaged sites to facilitate bone regeneration, is emphasized here. The review rationalizes that the choice of silk protein as a biomaterial is not only because of its natural polymeric nature, mechanical robustness, flexibility and wide range of cell compatibility but also because of its ability to template the growth of hydroxyapatite, the chief inorganic component of bone mineral matrix, resulting in improved osteointegration. The discussion extends to the role of inorganic ions such as Si and Ca as matrix components in combination with silk to influence bone regrowth. The effect of ions or growth factor-loaded vehicle incorporation into regenerative matrix, nanotopography is also considered.
Collapse
|
15
|
Font Tellado S, Bonani W, Balmayor ER, Foehr P, Motta A, Migliaresi C, van Griensven M. * Fabrication and Characterization of Biphasic Silk Fibroin Scaffolds for Tendon/Ligament-to-Bone Tissue Engineering. Tissue Eng Part A 2017; 23:859-872. [PMID: 28330431 DOI: 10.1089/ten.tea.2016.0460] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Tissue engineering is an attractive strategy for tendon/ligament-to-bone interface repair. The structure and extracellular matrix composition of the interface are complex and allow for a gradual mechanical stress transfer between tendons/ligaments and bone. Thus, scaffolds mimicking the structural features of the native interface may be able to better support functional tissue regeneration. In this study, we fabricated biphasic silk fibroin scaffolds designed to mimic the gradient in collagen molecule alignment present at the interface. The scaffolds had two different pore alignments: anisotropic at the tendon/ligament side and isotropic at the bone side. Total porosity ranged from 50% to 80% and the majority of pores (80-90%) were <100-300 μm. Young's modulus varied from 689 to 1322 kPa depending on the type of construct. In addition, human adipose-derived mesenchymal stem cells were cultured on the scaffolds to evaluate the effect of pore morphology on cell proliferation and gene expression. Biphasic scaffolds supported cell attachment and influenced cytoskeleton organization depending on pore alignment. In addition, the gene expression of tendon/ligament, enthesis, and cartilage markers significantly changed depending on pore alignment in each region of the scaffolds. In conclusion, the biphasic scaffolds fabricated in this study show promising features for tendon/ligament-to-bone tissue engineering.
Collapse
Affiliation(s)
- Sònia Font Tellado
- 1 Department of Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich , Munich, Germany
| | - Walter Bonani
- 2 Department of Industrial Engineering, BIOtech Research Center and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Trento , Trento, Italy .,3 Trento Research Unit, INSTM-National Interuniversity Consortium of Materials Science and Technology , Trento, Italy
| | - Elizabeth R Balmayor
- 1 Department of Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich , Munich, Germany
| | - Peter Foehr
- 4 Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich , Munich, Germany
| | - Antonella Motta
- 2 Department of Industrial Engineering, BIOtech Research Center and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Trento , Trento, Italy
| | - Claudio Migliaresi
- 2 Department of Industrial Engineering, BIOtech Research Center and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Trento , Trento, Italy .,3 Trento Research Unit, INSTM-National Interuniversity Consortium of Materials Science and Technology , Trento, Italy
| | - Martijn van Griensven
- 1 Department of Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich , Munich, Germany
| |
Collapse
|
16
|
Wang M, Li H, Si J, Dai J, Shi J, Wang X, Guo L, Shen G. Amniotic fluid-derived stem cells mixed with platelet rich plasma for restoration of rat alveolar bone defect. Acta Biochim Biophys Sin (Shanghai) 2017; 49:197-207. [PMID: 28104582 DOI: 10.1093/abbs/gmw133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Indexed: 12/12/2022] Open
Abstract
Stem cells isolated from the amniotic fluid have been shown as a promising candidate for cell therapy and tissue engineering. However, the experimental and preclinical applications of amniotic fluid-derived stem cells (AFSCs) in the very field of maxillofacial bone tissue engineering are still limited. In this study, rat AFSCs were successfully harvested and characterized in vitro. The rat AFSCs showed typical fibroblastoid morphology, stable proliferation activity and multi-differentiation potential. Flow-cytometry analysis demonstrated that these cells were positive for CD29, CD44, and CD90, while negative for hematopoietic markers such as CD34 and CD45. The regenerative performance of AFSCs-premixed with platelet rich plasma (PRP) gel in restoration of alveolar bone defect was further investigated using a modified rat maxillary alveolar defect model. Micro-computer tomography and histological examination showed a superior regenerative capacity of AFSCs-premixed with PRP gel at both 4 and 8 weeks after operation comparing with control groups. Moreover, the implanted AFSCs can survive in the defect site and directly participate in the bone tissue regeneration. Taken together, these results indicated the feasibility of an AFSCs-based alveolar bone tissue engineering strategy for alveolar defect restoration.
Collapse
Affiliation(s)
- Minjiao Wang
- Department of Oral and Craniomaxillofacial Science, Ninth People's Hospital College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Hongliang Li
- Department of Oral and Craniomaxillofacial Science, Ninth People's Hospital College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Jiawen Si
- Department of Oral and Craniomaxillofacial Science, Ninth People's Hospital College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Jiewen Dai
- Department of Oral and Craniomaxillofacial Science, Ninth People's Hospital College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Jun Shi
- Department of Oral and Craniomaxillofacial Science, Ninth People's Hospital College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Xudong Wang
- Department of Oral and Craniomaxillofacial Science, Ninth People's Hospital College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Lihe Guo
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guofang Shen
- Department of Oral and Craniomaxillofacial Science, Ninth People's Hospital College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| |
Collapse
|
17
|
Osteogenic Differentiation of Human Amniotic Fluid Mesenchymal Stem Cells Is Determined by Epigenetic Changes. Stem Cells Int 2016; 2016:6465307. [PMID: 27818691 PMCID: PMC5080506 DOI: 10.1155/2016/6465307] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 09/19/2016] [Indexed: 12/30/2022] Open
Abstract
Osteogenic differentiation of human amniotic fluid derived mesenchymal stem cells (AF-MSCs) has been widely studied in vitro and in vivo as a potential tool for regenerative medicine and tissue engineering. While most of the studies analyze changes in transcriptional profile during differentiation to date there is not much information regarding epigenetic changes in AF-MSCs during differentiation. The aim of our study was to evaluate epigenetic changes during osteogenic differentiation of AF-MS cells. Isolated AF-MSCs were characterized morphologically and osteogenic differentiation was confirmed by cell staining and determining expression of alkaline phosphatase and osteopontin by RT-qPCR. Variation in gene expression levels of pluripotency markers and specific microRNAs were also evaluated. Analysis of epigenetic changes revealed that levels of chromatin modifying enzymes such as Polycomb repressive complex 2 (PRC2) proteins (EZH2 and SUZ12), DNMT1, HDAC1, and HDAC2 were reduced after osteogenic differentiation of AF-MSCs. We demonstrated that the level of specific histone markers keeping active state of chromatin (H3K4me3, H3K9Ac, and others) increased and markers of repressed state of chromatin (H3K27me3) decreased. Our results show that osteogenic differentiation of AF-MSCs is conducted by various epigenetic alterations resulting in global chromatin remodeling and provide insights for further epigenetic investigations in human AF-MSCs.
Collapse
|
18
|
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
|
19
|
Allen AB, Zimmermann JA, Burnsed OA, Yakubovich DC, Stevens HY, Gazit Z, McDevitt TC, Guldberg RE. Environmental manipulation to promote stem cell survival in vivo: use of aggregation, oxygen carrier, and BMP-2 co-delivery strategies. J Mater Chem B 2016; 4:3594-3607. [DOI: 10.1039/c5tb02471d] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
While mesenchymal stem cell (MSC)-based strategies for critically-sized bone defect repair hold promise, poor cell survival in vivo remains a significant barrier to the translation of these therapeutics.
Collapse
Affiliation(s)
- Ashley B. Allen
- Wallace H. Coulter Department of Biomedical Engineering
- Parker H. Petit Institute for Bioengineering & Bioscience
- Georgia Institute of Technology
- Atlanta
- USA
| | - Josh A. Zimmermann
- Wallace H. Coulter Department of Biomedical Engineering
- Parker H. Petit Institute for Bioengineering & Bioscience
- Georgia Institute of Technology
- Atlanta
- USA
| | - Olivia A. Burnsed
- Wallace H. Coulter Department of Biomedical Engineering
- Parker H. Petit Institute for Bioengineering & Bioscience
- Georgia Institute of Technology
- Atlanta
- USA
| | - Doron Cohn Yakubovich
- Skeletal Biotech Laboratory
- The Hebrew University-Hadassah Faculty of Dental Medicine
- Jerusalem
- Israel
| | - Hazel Y. Stevens
- George W. Woodruff School of Mechanical Engineering
- Parker H. Petit Institute for Bioengineering & Bioscience
- Georgia Institute of Technology
- Atlanta
- USA
| | - Zulma Gazit
- Skeletal Biotech Laboratory
- The Hebrew University-Hadassah Faculty of Dental Medicine
- Jerusalem
- Israel
- Regenerative Medicine Institute
| | - Todd C. McDevitt
- Wallace H. Coulter Department of Biomedical Engineering
- Parker H. Petit Institute for Bioengineering & Bioscience
- Georgia Institute of Technology
- Atlanta
- USA
| | - Robert E. Guldberg
- George W. Woodruff School of Mechanical Engineering
- Parker H. Petit Institute for Bioengineering & Bioscience
- Georgia Institute of Technology
- Atlanta
- USA
| |
Collapse
|
20
|
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
|
21
|
Gholizadeh-Ghalehaziz S, Farahzadi R, Fathi E, Pashaiasl M. A Mini Overview of Isolation, Characterization and Application of Amniotic Fluid Stem Cells. Int J Stem Cells 2015; 8:115-120. [PMID: 26634059 PMCID: PMC4651275 DOI: 10.15283/ijsc.2015.8.2.115] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2015] [Indexed: 01/13/2023] Open
Abstract
Amniotic fluid represents rich sources of stem cells that can be used in treatments for a wide range of diseases. Amniotic fluid- stem cells have properties intermediate between embryonic and adult mesenchymal stem cells which make them particularly attractive for cellular regeneration and tissue engineering. Furthermore, scientists are interested in these cells because they come from the amniotic fluid that is routinely discarded after birth. In this review we give a brief introduction of amniotic fluid followed by a description of the cells present within this fluid and aim to summarize the all existing isolation methods, culturing, characterization and application of these cells. Finally, we elaborate on the differentiation and potential for these cells to promote regeneration of various tissue defects, including fetal tissue, the nervous system, heart, lungs, kidneys, bones, and cartilage in the form of table.
Collapse
Affiliation(s)
- Shiva Gholizadeh-Ghalehaziz
- Department of Molecular Medicine, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz,
Iran
| | - Raheleh Farahzadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz,
Iran
| | - Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz,
Iran
| | - Maryam Pashaiasl
- Department of Reproductive Medicine, Tabriz University of Medical Sciences, Tabriz,
Iran
| |
Collapse
|
22
|
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
|
23
|
Maraldi T, Beretti F, Guida M, Zavatti M, De Pol A. Role of hepatocyte growth factor in the immunomodulation potential of amniotic fluid stem cells. Stem Cells Transl Med 2015; 4:539-47. [PMID: 25873747 DOI: 10.5966/sctm.2014-0266] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/23/2015] [Indexed: 01/01/2023] Open
Abstract
UNLABELLED Human amniotic fluid stem cells (hAFSCs) may be useful for regenerative medicine because of their potential to differentiate into all three germ layers and to modulate immune response with different types of secretion molecules. This last issue has not been completely elucidated. The aim of this study was to investigate the secretome profile of the hAFSC, focusing on the role of hepatocyte growth factor (HGF) in immunoregulation through short and long cocultures with human peripheral blood mononuclear cells. We found that HGF produced by hAFSCs exerts a cytoprotective role, inducing an increase in caspase-dependent apoptosis in human immune cells. This study provides evidence supporting the hypothesis that amniotic fluid is an ideal source of stem cells for expansion and banking properties for therapeutic use. hAFSCs not only are less immunogenic but also can secrete immunoregulatory factors that may be useful in autoimmune diseases or allogenic implants. SIGNIFICANCE New information about the secretome pattern is reported in this paper. Human amniotic fluid stem cells (hAFSCs) possess immunomodulatory properties involving hepatocyte growth factor production. hAFSCs could be used in immunotherapies and might be able to avoid allogenic rejection.
Collapse
Affiliation(s)
- Tullia Maraldi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC) Research, Bolzano, Italy
| | - Francesca Beretti
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC) Research, Bolzano, Italy
| | - Marianna Guida
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC) Research, Bolzano, Italy
| | - Manuela Zavatti
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC) Research, Bolzano, Italy
| | - Anto De Pol
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC) Research, Bolzano, Italy
| |
Collapse
|
24
|
Comparative investigation of human amniotic epithelial cells and mesenchymal stem cells for application in bone tissue engineering. Stem Cells Int 2015; 2015:565732. [PMID: 25834575 PMCID: PMC4365333 DOI: 10.1155/2015/565732] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 12/01/2014] [Accepted: 12/01/2014] [Indexed: 02/07/2023] Open
Abstract
Emerging evidence suggests amniotic epithelial cells (AECs) as a promising source of progenitor cells in regenerative medicine and bone tissue engineering. However, investigations comparing the regenerative properties of AECs with other sources of stem cells are particularly needed before the feasibility of AECs in bone tissue engineering can be determined. This study aimed to compare human amniotic epithelial cells (hAECs), human bone marrow mesenchymal stem cells (hBMSCs), and human amniotic fluid derived mesenchymal stem cells (hAFMSCs) in terms of their morphology, proliferation, immunophenotype profile, and osteogenic capacity in vitro and in vivo. Not only greatly distinguished by cell morphology and proliferation, hAECs, hAFMSCs, and hBMSCs exhibited remarkably different signature regarding immunophenotypical profile. Microarray analysis revealed a different expression profile of genes involved in ossification along the three cell sources, highlighting the impact of different anatomical origin and molecular response to osteogenic induction on the final tissue-forming potential. Furthermore, our data indicated a potential role of FOXC2 in early osteogenic commitment.
Collapse
|
25
|
Resca E, Zavatti M, Maraldi T, Bertoni L, Beretti F, Guida M, La Sala G, Guillot P, David A, Sebire N, De Pol A, De Coppi P. Enrichment in c-Kit improved differentiation potential of amniotic membrane progenitor/stem cells. Placenta 2015; 36:18-26. [DOI: 10.1016/j.placenta.2014.11.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 10/31/2014] [Accepted: 11/03/2014] [Indexed: 12/15/2022]
|
26
|
Zavatti M, Bertoni L, Maraldi T, Resca E, Beretti F, Guida M, La Sala GB, De Pol A. Critical-size bone defect repair using amniotic fluid stem cell/collagen constructs: effect of oral ferutinin treatment in rats. Life Sci 2014; 121:174-83. [PMID: 25445219 DOI: 10.1016/j.lfs.2014.10.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 10/16/2014] [Accepted: 10/18/2014] [Indexed: 02/08/2023]
Abstract
AIMS This study aims to evaluate the bone regeneration in a rat calvarias critical size bone defect treated with a construct consisting of collagen type I and human amniotic fluid stem cells (AFSCs) after oral administration of phytoestrogen ferutinin. MAIN METHODS In 12 week old male rats (n=10), we performed two symmetric full-thickness cranial defects on each parietal region, and a scaffold was implanted into each cranial defect. The rats were divided into four groups: 1) collagen scaffold, 2) collagen scaffold+ferutinin at a dose of 2mg/kg/5 mL, 3) collagen scaffold + AFSCs, and 4) collagen scaffold + AFSCs + ferutinin. The rats were sacrificed after 4 weeks, and the calvariae were removed, fixed, embedded in paraffin and cut into 7 μm thick sections. Histomorphometric measures, immunohistochemical and immunofluorescence analyses were performed on the paraffin sections. KEY FINDINGS The histomorphometric analysis on H&E stained sections showed a significant increase in the regenerated area of the 4th group compared with the other groups. Immunohistochemistry performed with a human anti-mitochondrial antibody showed the presence of AFSCs 4 weeks after the transplant. Immunofluorescence analysis revealed the presence of osteocalcin and estrogen receptors (ERα and GPR30) in all groups, with a greater expression of all markers in samples where the scaffold was treated with AFSCs and the rats were orally administered ferutinin. SIGNIFICANCE Our results demonstrated that the oral administration of ferutinin is able to improve the bone regeneration of critical-size bone defects in vivo that is obtained with collagen-AFSCs constructs.
Collapse
Affiliation(s)
- Manuela 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.
| | - Laura 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
| | - Tullia 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
| | - Elisa Resca
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplants, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesca 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
| | | | - Giovanni B La Sala
- Unit of Obstetrics & Gynecology, IRCCS-Arcispedale Santa Maria Nuova, Reggio Emilia, Italy
| | - Anto 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
|
27
|
Pozzobon M, Piccoli M, De Coppi P. Stem cells from fetal membranes and amniotic fluid: markers for cell isolation and therapy. Cell Tissue Bank 2014; 15:199-211. [PMID: 24554400 DOI: 10.1007/s10561-014-9428-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 02/05/2014] [Indexed: 02/08/2023]
Abstract
Stem cell therapy is in constant need of new cell sources to conceive regenerative medicine approaches for diseases that are still without therapy. Scientists drew the attention toward amniotic membrane and amniotic fluid stem cells, since these sources possess many advantages: first of all as cells can be extracted from discarded foetal material it is inexpensive, secondly abundant stem cells can be obtained and finally, these stem cell sources are free from ethical considerations. Many studies have demonstrated the differentiation potential in vitro and in vivo toward mesenchymal and non-mesenchymal cell types; in addition the immune-modulatory properties make these cells a good candidate for allo- and xenotransplantation. This review offers an overview on markers characterisation and on the latest findings in pre-clinical or clinical setting of the stem cell populations isolated from these sources.
Collapse
Affiliation(s)
- Michela Pozzobon
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | | | | |
Collapse
|
28
|
Rossi F, Santoro M, Perale G. Polymeric scaffolds as stem cell carriers in bone repair. J Tissue Eng Regen Med 2013; 9:1093-119. [PMID: 24668819 DOI: 10.1002/term.1827] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 07/29/2013] [Accepted: 08/30/2013] [Indexed: 12/16/2022]
Affiliation(s)
- Filippo Rossi
- Department of Chemistry, Materials and Chemical Engineering; 'Giulio Natta' Politecnico di Milano; Milan Italy
| | - Marco Santoro
- Department of Chemical and Biomolecular Engineering; Rice University; Houston TX USA
| | - Giuseppe Perale
- Department of Chemistry, Materials and Chemical Engineering; 'Giulio Natta' Politecnico di Milano; Milan Italy
- Department of Innovative Technologies; University of Southern Switzerland; Manno Switzerland
- Swiss Institute for Regenerative Medicine; Taverne Switzerland
| |
Collapse
|
29
|
Carnevale G, Riccio M, Pisciotta A, Beretti F, Maraldi T, Zavatti M, Cavallini GM, La Sala GB, Ferrari A, De Pol A. In vitro differentiation into insulin-producing β-cells of stem cells isolated from human amniotic fluid and dental pulp. Dig Liver Dis 2013; 45:669-76. [PMID: 23643565 DOI: 10.1016/j.dld.2013.02.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Revised: 01/21/2013] [Accepted: 02/06/2013] [Indexed: 12/11/2022]
Abstract
AIM To investigate the ability of human amniotic fluid stem cells and human dental pulp stem cells to differentiate into insulin-producing cells. METHODS Human amniotic fluid stem cells and human dental pulp stem cells were induced to differentiate into pancreatic β-cells by a multistep protocol. Islet-like structures were assessed in differentiated human amniotic fluid stem cells and human dental pulp stem cells after 21 days of culture by dithizone staining. Pancreatic and duodenal homebox-1, insulin and Glut-2 expression were detected by immunofluorescence and confocal microscopy. Insulin secreted from differentiated cells was tested with SELDI-TOF MS and by enzyme-linked immunosorbent assay. RESULTS Human amniotic fluid stem cells and human dental pulp stem cells, after 7 days of differentiation started to form islet-like structures that became evident after 14 days of induction. SELDI-TOF MS analysis, revealed the presence of insulin in the media of differentiated cells at day 14, further confirmed by enzyme-linked immunosorbent assay after 7, 14 and 21 days. Both stem cell types expressed, after differentiation, pancreatic and duodenal homebox-1, insulin and Glut-2 and were positively stained by dithizone. Either the cytosol to nucleus translocation of pancreatic and duodenal homebox-1, either the expression of insulin, are regulated by glucose concentration changes. Day 21 islet-like structures derived from both human amniotic fluid stem cells and human dental pulp stem cell release insulin in a glucose-dependent manner. CONCLUSION The present study demonstrates the ability of human amniotic fluid stem cells and human dental pulp stem cell to differentiate into insulin-producing cells, offering a non-pancreatic, low-invasive source of cells for islet regeneration.
Collapse
Affiliation(s)
- Gianluca Carnevale
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Weber B, Kehl D, Bleul U, Behr L, Sammut S, Frese L, Ksiazek A, Achermann J, Stranzinger G, Robert J, Sanders B, Sidler M, Brokopp CE, Proulx ST, Frauenfelder T, Schoenauer R, Emmert MY, Falk V, Hoerstrup SP. In vitro fabrication of autologous living tissue-engineered vascular grafts based on prenatally harvested ovine amniotic fluid-derived stem cells. J Tissue Eng Regen Med 2013; 10:52-70. [PMID: 23881794 DOI: 10.1002/term.1781] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 03/19/2013] [Accepted: 04/22/2013] [Indexed: 12/11/2022]
Abstract
Amniotic fluid cells (AFCs) have been proposed as a valuable source for tissue engineering and regenerative medicine. However, before clinical implementation, rigorous evaluation of this cell source in clinically relevant animal models accepted by regulatory authorities is indispensable. Today, the ovine model represents one of the most accepted preclinical animal models, in particular for cardiovascular applications. Here, we investigate the isolation and use of autologous ovine AFCs as cell source for cardiovascular tissue engineering applications. Fetal fluids were aspirated in vivo from pregnant ewes (n = 9) and from explanted uteri post mortem at different gestational ages (n = 91). Amniotic non-allantoic fluid nature was evaluated biochemically and in vivo samples were compared with post mortem reference samples. Isolated cells revealed an immunohistochemical phenotype similar to ovine bone marrow-derived mesenchymal stem cells (MSCs) and showed expression of stem cell factors described for embryonic stem cells, such as NANOG and STAT-3. Isolated ovine amniotic fluid-derived MSCs were screened for numeric chromosomal aberrations and successfully differentiated into several mesodermal phenotypes. Myofibroblastic ovine AFC lineages were then successfully used for the in vitro fabrication of small- and large-diameter tissue-engineered vascular grafts (n = 10) and cardiovascular patches (n = 34), laying the foundation for the use of this relevant pre-clinical in vivo assessment model for future amniotic fluid cell-based therapeutic applications.
Collapse
Affiliation(s)
- Benedikt Weber
- Swiss Centre for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,Clinic for Cardiovascular Surgery and Department of Surgical Research, University Hospital of Zurich, Zurich, Switzerland
| | - Debora Kehl
- Swiss Centre for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,Clinic for Cardiovascular Surgery and Department of Surgical Research, University Hospital of Zurich, Zurich, Switzerland
| | - Ulrich Bleul
- Clinic of Reproductive Medicine, Department of Food Animals, Vetsuisse-Faculty University of Zurich, Zurich, Switzerland
| | - Luc Behr
- IMM Recherche, Institute Mutualiste Montsouris, Paris, France
| | | | - Laura Frese
- Swiss Centre for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,Clinic for Cardiovascular Surgery and Department of Surgical Research, University Hospital of Zurich, Zurich, Switzerland
| | - Agnieszka Ksiazek
- Swiss Centre for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,Clinic for Cardiovascular Surgery and Department of Surgical Research, University Hospital of Zurich, Zurich, Switzerland
| | | | - Gerald Stranzinger
- Breeding Biology Group, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Jérôme Robert
- Swiss Centre for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,Clinic for Cardiovascular Surgery and Department of Surgical Research, University Hospital of Zurich, Zurich, Switzerland.,Institute of Clinical Chemistry, University Hospital Zurich, Zurich, Switzerland
| | - Bart Sanders
- Department of Biomedical Engineering, Soft Tissue Biomechanics and Tissue Engineering, Eindhoven University of Technology, the Netherlands
| | - Michele Sidler
- Musculo-sceletal Research Unit, Vetsuisse-Faculty, University of Zurich, Zurich, Switzerland
| | - Chad E Brokopp
- Swiss Centre for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Steven T Proulx
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Switzerland
| | - Thomas Frauenfelder
- Department of Diagnostic Radiology, University Hospital Zurich, Zurich, Switzerland
| | - Roman Schoenauer
- Swiss Centre for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,Clinic for Cardiovascular Surgery and Department of Surgical Research, University Hospital of Zurich, Zurich, Switzerland
| | - Maximilian Y Emmert
- Swiss Centre for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,Clinic for Cardiovascular Surgery and Department of Surgical Research, University Hospital of Zurich, Zurich, Switzerland
| | - Volkmar Falk
- Swiss Centre for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,Clinic for Cardiovascular Surgery and Department of Surgical Research, University Hospital of Zurich, Zurich, Switzerland
| | - Simon P Hoerstrup
- Swiss Centre for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,Clinic for Cardiovascular Surgery and Department of Surgical Research, University Hospital of Zurich, Zurich, Switzerland.,Centre for Applied Biotechnology and Molecular Medicine (CABMM), Zurich, Switzerland
| |
Collapse
|
31
|
Resca E, Zavatti M, Bertoni L, Maraldi T, De Biasi S, Pisciotta A, Nicoli A, La Sala G, Guillot P, David A, Sebire N, De Coppi P, De Pol A. Enrichment in c-Kit+ enhances mesodermal and neural differentiation of human chorionic placental cells. Placenta 2013; 34:526-35. [DOI: 10.1016/j.placenta.2013.03.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 03/25/2013] [Accepted: 03/27/2013] [Indexed: 01/15/2023]
|
32
|
Maraldi T, Riccio M, Pisciotta A, Zavatti M, Carnevale G, Beretti F, La Sala GB, Motta A, De Pol A. Human amniotic fluid-derived and dental pulp-derived stem cells seeded into collagen scaffold repair critical-size bone defects promoting vascularization. Stem Cell Res Ther 2013; 4:53. [PMID: 23688855 PMCID: PMC3706961 DOI: 10.1186/scrt203] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 05/14/2013] [Indexed: 12/16/2022] Open
Abstract
Introduction The main aim of this study is to evaluate potential human stem cells, such as dental pulp stem cells and amniotic fluid stem cells, combined with collagen scaffold to reconstruct critical-size cranial bone defects in an animal model. Methods We performed two symmetric full-thickness cranial defects on each parietal region of rats and we replenished them with collagen scaffolds with or without stem cells already seeded into and addressed towards osteogenic lineage in vitro. After 4 and 8 weeks, cranial tissue samples were taken for histological and immunofluorescence analysis. Results We observed a new bone formation in all of the samples but the most relevant differences in defect correction were shown by stem cell–collagen samples 4 weeks after implant, suggesting a faster regeneration ability of the combined constructs. The presence of human cells in the newly formed bone was confirmed by confocal analysis with an antibody directed to a human mitochondrial protein. Furthermore, human cells were found to be an essential part of new vessel formation in the scaffold. Conclusion These data confirmed the strong potential of bioengineered constructs of stem cell–collagen scaffold for correcting large cranial defects in an animal model and highlighting the role of stem cells in neovascularization during skeletal defect reconstruction.
Collapse
|
33
|
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
|
34
|
Foss C, Migliaresi C, Motta A. The optimization of a scaffold for cartilage regeneration. Organogenesis 2013; 9:19-21. [PMID: 23538776 DOI: 10.4161/org.24257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Natural polymers offer various advantages in cartilage tissue engineering applications, thanks to their intrinsic bioactivity and adaptability, which can be exploited for the optimization of scaffold properties. In particular, silk fibroin has multifunctional features driven by the self-assembly of molecular subunits in appropriate environmental conditions. For these reasons, it was used in combination with hyaluronic acid to produce porous sponges for cartilage regeneration. The added amount of hyaluronic acid and the cross-linking with genipin modulated scaffold properties in a synergistic way, showing a strong inter-correlation among macroscopic and microscopic characteristics. Interestingly, hyaluronic acid affected silk fibroin conformation and induced a physical separation between the two material components in absence of genipin. Instead, this was prevented by the cross-linking reaction, resulting in a more interspersed network of protein and polysaccharide molecules partially resembling the structure of cartilage extracellular matrix. In addition, the systematic evaluation of sponge properties and how they can be modulated will represent a significant starting point for the interpretation of the complex outcomes driven by the scaffold in vitro and in vivo.
Collapse
Affiliation(s)
- Cristina Foss
- Department of Industrial Engineering and BIOtech Research Center, University of Trento, Mattarello, Trento, Italy
| | | | | |
Collapse
|
35
|
Pisciotta A, Riccio M, Carnevale G, Beretti F, Gibellini L, Maraldi T, Cavallini GM, Ferrari A, Bruzzesi G, De Pol A. Human serum promotes osteogenic differentiation of human dental pulp stem cells in vitro and in vivo. PLoS One 2012; 7:e50542. [PMID: 23209773 PMCID: PMC3510089 DOI: 10.1371/journal.pone.0050542] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 10/24/2012] [Indexed: 12/23/2022] Open
Abstract
Human dental pulp is a promising alternative source of stem cells for cell-based tissue engineering in regenerative medicine, for the easily recruitment with low invasivity for the patient and for the self-renewal and differentiation potential of cells. So far, in vitro culture of mesenchymal stem cells is usually based on supplementing culture and differentiation media with foetal calf serum (FCS). FCS is known to contain a great quantity of growth factors, and thus to promote cell attachment on plastic surface as well as expansion and differentiation. Nevertheless, FCS as an animal origin supplement may represent a potential means for disease transmission besides leading to a xenogenic immune response. Therefore, a significant interest is focused on investigating alternative supplements, in order to obtain a sufficient cell number for clinical application, avoiding the inconvenients of FCS use. In our study we have demonstrated that human serum (HS) is a suitable alternative to FCS, indeed its addition to culture medium induces a high hDPSCs proliferation rate and improves the in vitro osteogenic differentiation. Furthermore, hDPSCs-collagen constructs, pre-differentiated with HS-medium in vitro for 10 days, when implanted in immunocompromised rats, are able to restore critical size parietal bone defects. Therefore these data indicate that HS is a valid substitute for FCS to culture and differentiate in vitro hDPSCs in order to obtain a successful bone regeneration in vivo.
Collapse
Affiliation(s)
- Alessandra Pisciotta
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Riccio
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- * E-mail:
| | - Gianluca Carnevale
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesca Beretti
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Lara Gibellini
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Tullia Maraldi
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Gian Maria Cavallini
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Adriano Ferrari
- Department of Biomedical, Metabolic and Neuroscience, University of Modena and Reggio Emilia, Children Rehabilitation Special Unit, IRCCS Arcispedale Santa Maria Nuova, Reggio Emilia, Italy
| | | | - Anto De Pol
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
36
|
Veronesi F, Giavaresi G, Tschon M, Borsari V, Nicoli Aldini N, Fini M. Clinical use of bone marrow, bone marrow concentrate, and expanded bone marrow mesenchymal stem cells in cartilage disease. Stem Cells Dev 2012; 22:181-92. [PMID: 23030230 DOI: 10.1089/scd.2012.0373] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) from bone marrow (BM) are widely used for bone and less for cartilage tissue regeneration due to their self-renewal and differentiating properties into osteogenic or chondrogenic lineages. This review considers the last decade of clinical trials involving a two-step procedure, by expanding in vitro MSCs from BM, or the so called "one-step" procedure, using BM in toto or BM concentrate, for the regeneration of cartilage and osteochondral tissue defects. The following conclusions were drawn: (1) Cartilage defects that can be repaired by the two-step technique are about twice the size as those where the one-step method is used; (2) the two-step procedure is especially used for the treatment of osteoarthritic lesions, whereas the one-step procedure is used for osteochondral defects; (3) the number of transplanted cells ranges between 3.8×10(6) and 11.2×10(6) cells/mL, and the period of cell culture expansion of implanted MSCs varies widely with regard to the two-step procedure; (4) hyaluronic or collagenic scaffolds are used in all the clinical studies analyzed for both techniques; (5) the follow-up of the two-step procedure is longer than that of the one-step method, despite having a lower number of patients; and, finally, (6) the mean age of the patients (about 39 years old) is similar in both procedures. Clinical results underline the safety and good and encouraging outcomes for the use of MSCs in clinics. Although more standardized procedures are required, the length of follow-up and the number of patients observed should be augmented, and the design of trials should be implemented to achieve evidence-based results.
Collapse
Affiliation(s)
- Francesca Veronesi
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopedic Institute, Via Di Barbiano 1/10, Bologna, Italy
| | | | | | | | | | | |
Collapse
|
37
|
Bulj Z, Duchi S, Bevilacqua A, Gherardi A, Dozza B, Piccinini F, Adalgisa Mariani G, Lucarelli E, Giannini S, Donati D, Marmiroli S. Protein kinase B/AKT isoform 2 drives migration of human mesenchymal stem cells. Int J Oncol 2012; 42:118-26. [PMID: 23165443 PMCID: PMC3583637 DOI: 10.3892/ijo.2012.1700] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 10/08/2012] [Indexed: 01/01/2023] Open
Abstract
This study was designed to investigate the migratory behavior of adult human mesenchymal stem cells (MSC) and the underlying mechanism. Cell migration was assessed by transwell, wound healing and time-lapse in vivo motility assays. Pharmacological inhibitors were used to determine the potential mechanism responsible for cell migration and invasion. The tests that were implemented revealed that MSC were fairly migratory. Protein kinase B (AKT) was strongly activated at the basal level. Through our analyses we demonstrated that pharmacological inactivation of AKT2 but not AKT1 significantly decreased cell migration and invasion. Although preliminary, collectively our results indicate that AKT2 activation plays a critical role in enabling MSC migration.
Collapse
Affiliation(s)
- Zrinka Bulj
- Department of Biomedical Sciences, Cellular Signalling Laboratory, Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Cavallo C, Desando G, Columbaro M, Ferrari A, Zini N, Facchini A, Grigolo B. Chondrogenic differentiation of bone marrow concentrate grown onto a hylauronan scaffold: Rationale for its use in the treatment of cartilage lesions. J Biomed Mater Res A 2012; 101:1559-70. [DOI: 10.1002/jbm.a.34460] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 09/07/2012] [Accepted: 09/14/2012] [Indexed: 01/22/2023]
|
39
|
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
|
40
|
Amniotic fluid and amniotic membrane stem cells: marker discovery. Stem Cells Int 2012; 2012:107836. [PMID: 22701492 PMCID: PMC3372280 DOI: 10.1155/2012/107836] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 04/04/2012] [Indexed: 01/20/2023] Open
Abstract
Amniotic fluid (AF) and amniotic membrane (AM) have been recently characterized as promising sources of stem or progenitor cells. Both not only contain subpopulations with stem cell characteristics resembling to adult stem cells, such as mesenchymal stem cells, but also exhibit some embryonic stem cell properties like (i) expression of pluripotency markers, (ii) high expansion in vitro, or (iii) multilineage differentiation capacity. Recent efforts have been focused on the isolation and the detailed characterization of these stem cell types. However, variations in their phenotype, their heterogeneity described by different groups, and the absence of a single marker expressed only in these cells may prevent the isolation of a pure homogeneous stem cell population from these sources and their potential use of these cells in therapeutic applications. In this paper, we aim to summarize the recent progress in marker discovery for stem cells derived from fetal sources such as AF and AM, using novel methodologies based on transcriptomics, proteomics, or secretome analyses.
Collapse
|
41
|
Riccio M, Maraldi T, Pisciotta A, La Sala GB, Ferrari A, Bruzzesi G, Motta A, Migliaresi C, De Pol A. Fibroin scaffold repairs critical-size bone defects in vivo supported by human amniotic fluid and dental pulp stem cells. Tissue Eng Part A 2012; 18:1006-13. [PMID: 22166080 DOI: 10.1089/ten.tea.2011.0542] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The main aim of this study was the comparative evaluation of fibroin scaffolds combined with human stem cells, such as dental pulp stem cells (hDPSCs) and amniotic fluid stem cells (hAFSCs), used to repair critical-size cranial bone defects in immunocompromised rats. Two symmetric full-thickness cranial defects on each parietal region of rats have been replenished with silk fibroin scaffolds with or without preseeded stem cells addressed toward osteogenic lineage in vitro. Animals were euthanized after 4 weeks postoperatively and cranial tissue samples were taken for histological analysis. The presence of human cells in the new-formed bone was confirmed by confocal analysis with an antibody directed to a human mitochondrial protein. Fibroin scaffolds induced mature bone formation and defect correction, with higher bone amount produced by hAFSC-seeded scaffolds. Our findings demonstrated the strong potential of stem cells/fibroin bioengineered constructs for correcting large cranial defects in animal model and is likely a promising approach for the reconstruction of human large skeletal defects in craniofacial surgery.
Collapse
Affiliation(s)
- Massimo Riccio
- CEIA-Department of Laboratories, Pathological Anatomy and Forensic Medicine, University of Modena and Reggio Emilia, Modena, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|