1
|
Gantenbein B, Tang S, Guerrero J, Higuita-Castro N, Salazar-Puerta AI, Croft AS, Gazdhar A, Purmessur D. Non-viral Gene Delivery Methods for Bone and Joints. Front Bioeng Biotechnol 2020; 8:598466. [PMID: 33330428 PMCID: PMC7711090 DOI: 10.3389/fbioe.2020.598466] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/26/2020] [Indexed: 12/12/2022] Open
Abstract
Viral carrier transport efficiency of gene delivery is high, depending on the type of vector. However, viral delivery poses significant safety concerns such as inefficient/unpredictable reprogramming outcomes, genomic integration, as well as unwarranted immune responses and toxicity. Thus, non-viral gene delivery methods are more feasible for translation as these allow safer delivery of genes and can modulate gene expression transiently both in vivo, ex vivo, and in vitro. Based on current studies, the efficiency of these technologies appears to be more limited, but they are appealing for clinical translation. This review presents a summary of recent advancements in orthopedics, where primarily bone and joints from the musculoskeletal apparatus were targeted. In connective tissues, which are known to have a poor healing capacity, and have a relatively low cell-density, i.e., articular cartilage, bone, and the intervertebral disk (IVD) several approaches have recently been undertaken. We provide a brief overview of the existing technologies, using nano-spheres/engineered vesicles, lipofection, and in vivo electroporation. Here, delivery for microRNA (miRNA), and silencing RNA (siRNA) and DNA plasmids will be discussed. Recent studies will be summarized that aimed to improve regeneration of these tissues, involving the delivery of bone morphogenic proteins (BMPs), such as BMP2 for improvement of bone healing. For articular cartilage/osteochondral junction, non-viral methods concentrate on targeted delivery to chondrocytes or MSCs for tissue engineering-based approaches. For the IVD, growth factors such as GDF5 or GDF6 or developmental transcription factors such as Brachyury or FOXF1 seem to be of high clinical interest. However, the most efficient method of gene transfer is still elusive, as several preclinical studies have reported many different non-viral methods and clinical translation of these techniques still needs to be validated. Here we discuss the non-viral methods applied for bone and joint and propose methods that can be promising in clinical use.
Collapse
Affiliation(s)
- Benjamin Gantenbein
- Tissue Engineering for Orthopaedics and Mechanobiology, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Shirley Tang
- Department of Biomedical Engineering and Department of Orthopaedics, Spine Research Institute Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Julien Guerrero
- Tissue Engineering for Orthopaedics and Mechanobiology, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Natalia Higuita-Castro
- Department of Biomedical Engineering and Department of Surgery, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Ana I Salazar-Puerta
- Department of Biomedical Engineering and Department of Surgery, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Andreas S Croft
- Tissue Engineering for Orthopaedics and Mechanobiology, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Amiq Gazdhar
- Department of Pulmonary Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| | - Devina Purmessur
- Department of Biomedical Engineering and Department of Orthopaedics, Spine Research Institute Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
2
|
The fabrication of biomimetic biphasic CAN-PAC hydrogel with a seamless interfacial layer applied in osteochondral defect repair. Bone Res 2017; 5:17018. [PMID: 28698817 PMCID: PMC5496380 DOI: 10.1038/boneres.2017.18] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 01/23/2017] [Accepted: 02/23/2017] [Indexed: 02/05/2023] Open
Abstract
Cartilage tissue engineering based on biomimetic scaffolds has become a rapidly developing strategy for repairing cartilage defects. In this study, a biphasic CAN-PAC hydrogel for osteochondral defect (OCD) regeneration was fabricated based on the density difference between the two layers via a thermally reactive, rapid cross-linking method. The upper hydrogel was cross-linked by CSMA and NIPAm, and the lower hydrogel was composed of PECDA, AAm and PEGDA. The interface between the two layers was first grafted by the physical cross-linking of calcium gluconate and alginate, followed by the chemical cross-linking of the carbon-carbon double bonds in the other components. The pore sizes of the upper and lower hydrogels were ~187.4 and ~112.6 μm, respectively. The moduli of the upper and lower hydrogels were ~0.065 and ~0.261 MPa. This prepared bilayer hydrogel exhibited the characteristics of mimetic composition, mimetic structure and mimetic stiffness, which provided a microenvironment for sustaining cell attachment and viability. Meanwhile, the biodegradability and biocompatibility of the CAN-PAC hydrogel were examined in vivo. Furthermore, an osteochondral defect model was developed in rabbits, and the bilayer hydrogels were implanted into the defect. The regenerated tissues in the bilayer hydrogel group exhibited new translucent cartilage and repaired subchondral bone, indicating that the hydrogel can enhance the repair of osteochondral defects.
Collapse
|
3
|
Im GI. Regeneration of articular cartilage using adipose stem cells. J Biomed Mater Res A 2016; 104:1830-44. [PMID: 26990234 DOI: 10.1002/jbm.a.35705] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 03/02/2016] [Indexed: 12/16/2022]
Abstract
Articular cartilage (AC) has limited potential for self-regeneration and damage to AC eventually leads to the development and progression of osteoarthritis (OA). Cell implantation strategies have emerged as a new treatment modality to regenerate AC. Adipose stem cells/adipose-derived stromal cells (ASCs) have gained attention due to their abundance, excellent proliferative potential, and minimal morbidity during harvest. These advantages lower the cost of cell therapy by circumventing time-consuming procedure of culture expansion. ASCs have drawn attention as a potential source for cartilage regeneration since the feasibility of chondrogenesis from ASCs was first reported. After several groups reported inferior chondrogenesis from ASCs, numerous methods were devised to overcome the intrinsic properties. Most in vivo animal studies have reported good results using predifferentiated or undifferentiated, autologous or allogeneic ASCs to regenerate cartilage in osteochondral defects or surgically-induced OA. In this review, we summarize literature on the isolation and in vitro differentiation processes of ASCs, in vivo studies to regenerate AC in osteochondral defects and OA using ASCs, and clinical applications of ASCs. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 1830-1844, 2016.
Collapse
Affiliation(s)
- Gun-Il Im
- Department of Orthopedics, Dongguk University Ilsan Hospital, Goyang, Korea
| |
Collapse
|
4
|
Wu L, Cai X, Zhang S, Karperien M, Lin Y. Regeneration of articular cartilage by adipose tissue derived mesenchymal stem cells: perspectives from stem cell biology and molecular medicine. J Cell Physiol 2013; 228:938-44. [PMID: 23042088 DOI: 10.1002/jcp.24255] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2012] [Accepted: 09/27/2012] [Indexed: 01/03/2023]
Abstract
Adipose-derived stem cells (ASCs) have been discovered for more than a decade. Due to the large numbers of cells that can be harvested with relatively little donor morbidity, they are considered to be an attractive alternative to bone marrow derived mesenchymal stem cells. Consequently, isolation and differentiation of ASCs draw great attention in the research of tissue engineering and regenerative medicine. Cartilage defects cause big therapeutic problems because of their low self-repair capacity. Application of ASCs in cartilage regeneration gives hope to treat cartilage defects with autologous stem cells. In recent years, a lot of studies have been performed to test the possibility of using ASCs to re-construct damaged cartilage tissue. In this article, we have reviewed the most up-to-date articles utilizing ASCs for cartilage regeneration in basic and translational research. Our topic covers differentiation of adipose tissue derived mesenchymal stem cells into chondrocytes, increased cartilage formation by co-culture of ASCs with chondrocytes and enhancing chondrogenic differentiation of ASCs by gene manipulation.
Collapse
Affiliation(s)
- Ling Wu
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, P.R. China
| | | | | | | | | |
Collapse
|
5
|
Xiao J, Yang X, Jing W, Guo W, Sun Q, Lin Y, Liu L, Meng W, Tian W. Adipogenic and osteogenic differentiation of Lin(-)CD271(+)Sca-1(+) adipose-derived stem cells. Mol Cell Biochem 2013; 377:107-19. [PMID: 23430356 DOI: 10.1007/s11010-013-1575-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 01/24/2013] [Indexed: 12/29/2022]
Abstract
Adipose-derived stem cells (ASCs) have been defined as cells that undergo sustained in vitro growth and have multilineage differentiation potential. However, the identity and purification of ASCs has proved elusive due to the lack of specific markers and poor understanding of their physiological roles. Here, we prospectively isolated and identified a restricted homogeneous subpopulation of ASCs (Lin(-)CD271(+)Sca-1(+)) from mouse adipose tissues on the basis of cell-surface markers. Individual ASCs generated colony-forming unit-fibroblast at a high frequency and could differentiate into adipocytes, osteoblasts, and chondrocytes in vitro. Expansion of ASCs in a large quantity was feasible in medium supplemented with fibroblast growth factor-2 and leukemia inhibitory factor, without loss of adipogenic and osteogenic differentiation capacity. Moreover, we found that the transplanted ASCs can differentiate into adipocytes in adipogenic microenvironment in vivo and osteoblasts in osteogenic microenvironment in vivo. Thus we proved that Lin, CD271, and Sca-1 could be used as the specific markers to purify ASCs from adipose tissue. The method we established to identify ASCs as defined in vivo entities will help develop ASCs transplantation as a new therapeutic strategy for bone regeneration and adipose tissue regeneration in clinic.
Collapse
Affiliation(s)
- Jingang Xiao
- State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
| | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Bluguermann C, Wu L, Petrigliano F, McAllister D, Miriuka S, Evseenko DA. Novel aspects of parenchymal-mesenchymal interactions: from cell types to molecules and beyond. Cell Biochem Funct 2013; 31:271-80. [PMID: 23315627 DOI: 10.1002/cbf.2950] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 12/06/2012] [Accepted: 12/06/2012] [Indexed: 12/26/2022]
Abstract
Mesenchymal stem or stromal cells (MSCs) were initially isolated from the bone marrow and received their name on the basis of their ability to differentiate into multiple lineages such as bone, cartilage, fat and muscle. However, more recent studies suggest that MSCs residing in perivascular compartments of the small and large blood vessels play a regulatory function supporting physiologic and pathologic responses of parenchymal cells, which define the functional representation of an organ or tissue. MSCs secrete or express factors that reach neighbouring parenchymal cells via either a paracrine effect or a direct cell-to-cell interaction promoting functional activity, survival and proliferation of the parenchymal cells. Previous concept of 'epithelial-stromal' interactions can now be widened. Given that MSC can also support hematopoietic, neuronal and other non-epithelial parenchymal lineages, terms 'parenchymal-stromal' or 'parenchymal-mesenchymal' interactions may better describe the supportive or 'trophic' functions of MSC. Importantly, in many cases, MSCs specifically provide supportive microenvironment for the most primitive stem or progenitor populations and therefore can play a role as 'stem/progenitor niche' forming cells. So far, regulatory roles of MSCs have been reported in many tissues. In this review article, we summarize the latest studies that focused on the supportive function of MSC. This thread of research leads to a new perspective on the interactions between parenchymal and mesenchymal cells and justifies a principally novel approach for regenerative medicine based on co-application of MSC and parenchymal cell for the most efficient tissue repair.
Collapse
Affiliation(s)
- Carolina Bluguermann
- Department of Orthopaedic Surgery, Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California at Los Angeles (UCLA), Los Angeles, CA, USA
| | | | | | | | | | | |
Collapse
|
7
|
Raabe O, Reich C, Wenisch S, Hild A, Burg-Roderfeld M, Siebert HC, Arnhold S. Hydrolyzed fish collagen induced chondrogenic differentiation of equine adipose tissue-derived stromal cells. Histochem Cell Biol 2010; 134:545-54. [PMID: 21076963 DOI: 10.1007/s00418-010-0760-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2010] [Indexed: 10/18/2022]
Abstract
Adipose-derived stromal cells (ADSCs) are multipotent cells which, in the presence of appropriate stimuli, can differentiate into various lineages such as the osteogenic, adipogenic and chondrogenic. In this study, we investigated the effect of transforming growth factor beta 1 (TGF-β1) in comparison to hydrolyzed fish collagen in terms of the chondrogenic differentiation potential of ADSCs. ADSCs were isolated from subcutaneous fat of horses by liposuction. Chondrogenesis was investigated using a pellet culture system. The differentiation medium was either supplemented with TGF-β1 (5 ng/ml) or fish collagen (0.5 mg/ml) for a 3 week period. After the 3 weeks in vitro differentiation, RT-PCR and histological staining for proteoglycan synthesis and type II collagen were performed to evaluate the degree of chondrogenic differentiation and the formation of cartilaginous extracellular matrix (ECM). The differentiation of ADSCs induced by TGF-β1 showed a high expression of glycosaminoglycan (GAG). Histological analysis of cultures stimulated by hydrolyzed fish collagen demonstrated an even higher GAG expression than cultures stimulated under standard conditions by TGF-β1. The expression of cartilage-specific type II collagen and Sox9 was about the same in both stimulated cultures. In this study, chondrogenesis was as effectively induced by hydrolyzed fish collagen as it was successfully induced by TGF-β1. These findings demonstrated that hydrolyzed fish collagen alone has the potential to induce and maintain ADSCs-derived chondrogenesis. These results support the application of ADSCs in equine veterinary tissue engineering, especially for cartilage repair.
Collapse
Affiliation(s)
- O Raabe
- Institute of Veterinary Anatomy, Histology, and Embryology, Justus-Liebig University of Giessen, Giessen, Germany.
| | | | | | | | | | | | | |
Collapse
|
8
|
Multilineage differentiation of muscle-derived stem cells from GFP transgenic mice. Biotechnol Lett 2010; 32:1745-52. [PMID: 20632072 DOI: 10.1007/s10529-010-0344-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2010] [Accepted: 06/28/2010] [Indexed: 10/19/2022]
Abstract
It is unclear whether green fluorescent protein (GFP) expression is maintained during the course of multilineage differentiation of muscle-derived stem cells (MDSCs). We isolated MDSCs from GFP-transgenic mice and transferred them to chondrogenic, neurogenic or myogenic media. Multilineage differentiation was examined by morphological observation, histological staining, immunocytochemical staining, real-time RT-PCR and Western blot. Both differentiated cells and non-differentiated cells maintained stable GFP expression until the cells exhibited a senescent phenotype. Thus, MDSCs from GFP-transgenic mice have multilineage potential in vitro and that GFP expression does not influence the multilineage potential of MDSCs (or vice versa).
Collapse
|
9
|
Miot S, Gianni-Barrera R, Pelttari K, Acharya C, Mainil-Varlet P, Juelke H, Jaquiery C, Candrian C, Barbero A, Martin I. In vitro and in vivo validation of human and goat chondrocyte labeling by green fluorescent protein lentivirus transduction. Tissue Eng Part C Methods 2010; 16:11-21. [PMID: 19327004 DOI: 10.1089/ten.tec.2008.0698] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
We investigated whether human articular chondrocytes can be labeled efficiently and for long-term with a green fluorescent protein (GFP) lentivirus and whether the viral transduction would influence cell proliferation and tissue-forming capacity. The method was then applied to track goat articular chondrocytes after autologous implantation in cartilage defects. Expression of GFP in transduced chondrocytes was detected cytofluorimetrically and immunohistochemically. Chondrogenic capacity of chondrocytes was assessed by Safranin-O staining, immunostaining for type II collagen, and glycosaminoglycan content. Human articular chondrocytes were efficiently transduced with GFP lentivirus (73.4 +/- 0.5% at passage 1) and maintained the expression of GFP up to 22 weeks of in vitro culture after transduction. Upon implantation in nude mice, 12 weeks after transduction, the percentage of labeled cells (73.6 +/- 3.3%) was similar to the initial one. Importantly, viral transduction of chondrocytes did not affect the cell proliferation rate, chondrogenic differentiation, or tissue-forming capacity, either in vitro or in vivo. Goat articular chondrocytes were also efficiently transduced with GFP lentivirus (78.3 +/- 3.2%) and maintained the expression of GFP in the reparative tissue after orthotopic implantation. This study demonstrates the feasibility of efficient and relatively long-term labeling of human chondrocytes for co-culture on integration studies, and indicates the potential of this stable labeling technique for tracking animal chondrocytes for in cartilage repair studies.
Collapse
Affiliation(s)
- Sylvie Miot
- Department of Surgery and of Biomedicine, University Hospital Basel , Basel, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
In vitro osteogenic differentiation of adipose stem cells after lentiviral transduction with green fluorescent protein. J Craniofac Surg 2010; 20:2193-9. [PMID: 19934675 DOI: 10.1097/scs.0b013e3181bf04af] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Adipose-derived stem cells (ASCs) have the potential to differentiate into osteogenic cells that can be seeded into scaffolds for tissue engineering for use in craniofacial bone defects. Green fluorescent protein (GFP) has been widely used as a lineage marker for mammalian cells. The use of fluorescent proteins enables cells to be tracked during manipulation such as osteogenic differentiation within three-dimensional scaffolds. The purpose of this study was to examine whether ASCs introduced with GFP-encoding lentivirus vector exhibit adequate GFP fluorescence and whether the expression of GFP interfered with osteogenic differentiation of ASCs in both monolayer and three-dimensional scaffolds in vitro. METHODS Primary ASCs were harvested from the inguinal fat pad of Sprague Dawley rats. Isolated ASCs were cultured and infected with a lentiviral vector encoding GFP and plated into both monolayers and three-dimensional scaffolds in vitro. The cells were then placed in osteogenic medium. Osteogenic differentiation of the GFP-ASCs was assessed using alizarin red S, alkaline phosphate staining, and immunohistochemistry staining of osteocalcin with quantification of alizarin red S and osteocalcin staining. RESULTS The efficacy of infection of ASCs with a lentiviral vector encoding GFP was high. Cell-cultured GFP-ASCs remained fluorescent over the 8 weeks of the study period. The GFP-ASCs were successfully induced into osteogenic cells both in monolayers and three-dimensional scaffolds. Whereas the quanitification of alizarin red S revealed no difference between osteoinduced ASCs with or without GFP, the quantification of osteocalcin revealed increased staining in the GFP group. CONCLUSIONS Transduction of isolated ASCs using a lentiviral vector encoding GFP is an effective method for tracing osteoinduced ASCs in vitro. Quantification data showed no decrease in staining of the osteoinduced ASCs.
Collapse
|
11
|
Kishi K, Imanishi N, Ohara H, Ninomiya R, Okabe K, Hattori N, Kubota Y, Nakajima H, Nakajima T. Distribution of adipose-derived stem cells in adipose tissues from human cadavers. J Plast Reconstr Aesthet Surg 2009; 63:1717-22. [PMID: 20044319 DOI: 10.1016/j.bjps.2009.10.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2008] [Revised: 09/06/2009] [Accepted: 10/22/2009] [Indexed: 10/20/2022]
Abstract
BACKGROUND Adipose-derived stem cells (ASCs) possess multipotency in vivo and in vitro, and thus are thought to be very promising precursors for use in regenerative medicine. ASCs can be concentrated from adipose tissue by enzymatic digestion and transplanted to increase angiogenesis or for cosmesis. ASC transplants are now being performed in a clinical setting. Although data on ASCs are extensive, the distribution of ASCs in human fat tissue has not been fully clarified. Thus, it is important to identify the distribution of ASCs to obtain cell populations rich in ASCs for clinical use. METHODS ASCs express CD34, a cell surface marker. As CD34 is also expressed by endothelial cells, we immunohistochemically stained 2-μm-thick serial paraffin sections of fat tissue obtained from various parts of formalin-fixed cadavers with anti-CD31 and anti-CD34 antibodies to distinguish ASCs from endothelial cells. RESULTS CD34(+)/CD31(-) cells were mainly found in connective tissue tracts and perivascularly. Among fat tissues obtained from various sites, fat tissues in the thoracic back and lower abdomen were richest in CD34(+)/CD31(-) cells. CONCLUSION The concentrations of CD34(+)/CD31(-) cells in adipose tissues differ between sites. The sites most highly enriched for ASCs were identified, and it is now possible to select the best sites for collection of ASCs for transplantation.
Collapse
Affiliation(s)
- Kazuo Kishi
- Department of Plastic and Reconstructive Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku Tokyo, 160-8582, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Qin Y, Ji H, Wu Y, Liu H. Chromosomal instability of murine adipose tissue-derived mesenchymal stem cells in long-term culture and development of cloned embryos. CLONING AND STEM CELLS 2009; 11:445-52. [PMID: 19594392 DOI: 10.1089/clo.2009.0006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Mice are the most commonly used laboratory animals for research, and some mouse stem cells, such as induced pluripotent stem cells, embryonic stem cells, and mesenchymal stem cells (MSCs), are also widely used in basic research. It is thus important to know if these stem cells maintain their genomic stability when cultured. Murine bone marrow-derived mesenchymal stem cells (BMSCs) appear to undergo spontaneous transformation in vitro. Murine adipose tissue-derived mesenchymal stem cells (ADSCs), like BMSCs, have the potential to differentiate into multiple lineages. In this study, we used G-banding, induction of multiple-lineage differentiation, flow cytometry, and nuclear transfer (NT), and found that murine ADSCs also displayed chromosomal instability in long-term culture. Furthermore, we performed NT using murine ADSCs to study the nuclear reprogramming ability of undifferentiated adult stem cells and to find a new efficient donor for NT. Using the stem cells did not increase the percentage of NT embryos that developed to the morula/blastocyst stage, compared with cloned embryos from cumulus cells. This may be because the stem cells displayed chromosomal instability. This is the first reported study of the use of ADSCs for NT in mice. ADSCs could provide an alternative donor cell type for NT in other species, with the advantages of easy harvesting involving little or no pain or trauma.
Collapse
Affiliation(s)
- Yiren Qin
- Department of Histology and Embryology, Harbin Medical University, Harbin, PR China
| | | | | | | |
Collapse
|
13
|
Wu L, Zhu F, Wu Y, Lin Y, Nie X, Jing W, Qiao J, Liu L, Tang W, Zheng X, Tian W. Dentin Sialophosphoprotein-Promoted Mineralization and Expression of Odontogenic Genes in Adipose-Derived Stromal Cells. Cells Tissues Organs 2008; 187:103-12. [DOI: 10.1159/000110079] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2007] [Indexed: 01/06/2023] Open
|
14
|
Bone regeneration by BMP-2 enhanced adipose stem cells loading on alginate gel. Histochem Cell Biol 2007; 129:203-10. [DOI: 10.1007/s00418-007-0351-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2007] [Indexed: 12/13/2022]
|
15
|
Ectopic adipogenesis of preconditioned adipose-derived stromal cells in an alginate system. Cell Tissue Res 2007; 330:567-72. [PMID: 17922143 DOI: 10.1007/s00441-007-0493-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2007] [Accepted: 08/01/2007] [Indexed: 10/22/2022]
Abstract
Soft tissue substitutes are increasingly needed in reconstructive and plastic surgery, although difficulties arise when autografts or allografts have to provide long-term satisfactory replacement in soft tissue restoration. Adipose tissue engineering has highlighted the possibility of soft tissue restoration by using regeneration strategies. As few reports exist regarding adipose regeneration with alginate systems mixed with adipose-derived stromal cells (ASCs), the present study has attempted to evaluate the adipogenic differentiation of ASCs isolated from BALB/c mice in vitro and to define the potential of ASCs to form ectopic adipose tissue in an alginate system with subcutaneous implantation in nude mice. After 10 days induction in vitro, prediferentiated ASCs were seeded in an alginate gel system and implanted subcutaneously into the dorsum of nude mice. Adipogenesis of the newly formed tissue at 8 weeks post-implantation was confirmed by oil red O staining, reverse transcription/polymerase chain reaction, and Western blot analysis. This study thus provides evidence that the alginate system has the ability to form ectopic adipose tissue when mixed with predifferentiated ASCs. The results also supply clues for improving adipose tissue engineering and soft tissue defects repair in adults.
Collapse
|
16
|
Lin Y, Wang T, Wu L, Jing W, Chen X, Li Z, Liu L, Tang W, Zheng X, Tian W. Ectopic and in situ bone formation of adipose tissue-derived stromal cells in biphasic calcium phosphate nanocomposite. J Biomed Mater Res A 2007; 81:900-10. [PMID: 17236222 DOI: 10.1002/jbm.a.31149] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Adipose-derived stromal cells (ASCs) have the potential to differentiate into a variety of cell lineages both in vitro and in vivo. A novel biodegradable biphasic calcium phosphate nanocomposite (NanoBCP) comprising beta-tricalcium phosphate matrix and hydroxyl apatite nanofibers is favorable for bone tissue engineering. In this study, ASCs were harvested from Sprague-Dawley (SD) rats and induced to osteogenesis before seeded into porous NanoBCP scaffold. To determine ectopic in vivo osteogenic differentiation, these constructs were implanted in nude mice subcutaneously. Meanwhile, the ability of engineered constructs to stimulate in situ bone repair was assessed in rat critical-size cranial defects. The defects were filled with NanoBCP containing osteogenic ASCs in experimental group; with cell-free NanoBCP in negative controls; and with nothing in blank controls. The retrieved specimens were analyzed with morphological, histological, and molecular methods. Histological analysis of the retrieved specimens from nude mice in experimental group showed obvious ectopic bone formation. There were positive expression of osteopontin (OPN) and osteocalcin (OCN) at RNA and protein level. As for the cranial defects, there was complete repair in experimental group, but only partial repair in negative controls. The radiographs, H&E staining, and Masson's trichrome method showed better bone regeneration at experimental sites. Combining osteogenic ASCs with NanoBCP can lead to formation of ectopic new bone. Furthermore, the approach can also stimulate bone regeneration and repair for the large size bone defects.
Collapse
Affiliation(s)
- Yunfeng Lin
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Li ZY, Chen L, Liu L, Lin YF, Li SW, Tian WD. Odontogenic potential of bone marrow mesenchymal stem cells. J Oral Maxillofac Surg 2007; 65:494-500. [PMID: 17307598 DOI: 10.1016/j.joms.2006.09.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2005] [Revised: 07/22/2006] [Accepted: 09/28/2006] [Indexed: 12/30/2022]
Abstract
PURPOSE This study aimed to investigate the odontogenic potential of bone marrow mesenchymal stem cells (BM-MSCs) for seeding in tooth regeneration. MATERIALS AND METHODS In this study, BM-MSCs were co-cultured with oral epithelial cells derived from rat embryos. Expression of the odontogenic genes Pax9, DMP1, and DSPP was detected by the reverse-transcription polymerase chain reaction (RT-PCR) technique. To further characterize the odontogenic potential of BM-MSCs, the gold standard in vivo transplantation system was used. RESULTS The results revealed that Pax9, DMP1, and DSPP expression was detected by RT-PCR only after co-culture of BM-MSCs and oral epithelial cells derived from embryos age E11.5. Histological analyses of the BM-MSCs/epithelial cell mass demonstrated the presence of tooth-like structures. CONCLUSIONS The series of experiments both in vitro and in vivo demonstrated that BM-MSCs can differentiate into functional odontoblast-like cells. This implies that BM-MSCs may become a novel source of cells for seeding in tooth regeneration research.
Collapse
Affiliation(s)
- Zhi-Yong Li
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | | | | | | | | | | |
Collapse
|
18
|
Wu L, Wu Y, Lin Y, Jing W, Nie X, Qiao J, Liu L, Tang W, Tian W. Osteogenic differentiation of adipose derived stem cells promoted by overexpression of osterix. Mol Cell Biochem 2007; 301:83-92. [PMID: 17206379 DOI: 10.1007/s11010-006-9399-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2006] [Accepted: 12/06/2006] [Indexed: 12/21/2022]
Abstract
Adipose-derived stem cells (ASCs) are considered to be multipotent mesenchymal stem cells that are easily induced to differentiate into functional osteoblasts both in vitro and in vivo. Osterix (Osx) is a zinc finger-containing transcription factor of Sp gene family, which plays important roles in bone development and mineralization. In this study, we hypothesized that overexpression of Osx in murine ASCs would promote their osteogenic differentiation in vitro. A plasmid expressing Osx (pcDNA3.1-Osx) was constructed and applied to transfect monolayers of murine ASCs. Then expression of bone-related genes, nodule formation, proliferation rate, and alkaline phosphatase activity were examined to evaluate the osteogenic potential of ASCs with pcDNA3.1-Osx transfection. Results of RT-PCR and immunohistochemistry showed that pcDNA3.1-Osx transfection enhanced the expression of bone matrix proteins, such as bone sialoprotein, osteocalcin, osteopontin, and Collagen type I in ASCs. At the same time, overexpression of Osx in ASCs enhanced alkaline phosphatase activity and capability to form mineralized nodules, while not inhibited their proliferation rate. These results indicated that pcDNA3.1-Osx transfection promoted the osteogenic differentiation of ASCs, while not affecting their proliferative ability. Since they can be easily isolated and genetically modified, ASCs are hopeful cell sources in the further application of hard tissue engineering.
Collapse
Affiliation(s)
- Ling Wu
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Lin Y, Liu L, Li Z, Qiao J, Wu L, Tang W, Zheng X, Chen X, Yan Z, Tian W. Pluripotency potential of human adipose-derived stem cells marked with exogenous green fluorescent protein. Mol Cell Biochem 2006; 291:1-10. [PMID: 16718363 DOI: 10.1007/s11010-006-9188-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2006] [Accepted: 03/08/2006] [Indexed: 12/17/2022]
Abstract
Musculoskeletal tissues regeneration requires rapid expansion of seeding cells both in vitro and in vivo while maintaining their multilineage differentiation ability. Human adipose-derived stem cells (ASCs) are considered to contain multipotent mesenchymal stem cells. Monolayer cultures of human ASCs were isolated from human lipoaspirates and passaged 3 times and then infected with replication-incompetent adenoviral vectors carrying green fluorescent protein (Ad/GFP) genes. Then, Ad/GFP infected human ASCs were transferred to osteogenic, chondrogenic, adipogenic, and myogenic medium. The morphological characterization of induced cells was observed using phase-contrast microscopy and fluorescence microscopy. The expression of marker proteins or genes was measured by immunocytochemical and RT-PCR analysis. Osteopontin (OPN), and osteocalcin (OCN) were positive in osteogenic lineages, aggrecan and SOX9 were positive in chondrogenic ones, peroxisome proliferator-activated receptor (PPAR-gamma2) and lipoprotein lipase (LPL) were positive in adipogenic ones, and myogenin and myod1 was positive in myogenic ones. At the same time, the results of fluorescence microscopic imaging proved that the high level of GFP expression during ASCs differentiation maintained stable nearly 2 months. So the exogenous GFP and multilineage potential of human ASCs had no severe influences on each other. Since the human ASCs can be easily obtained and abundant, it is proposed that they may be promising candidate cells for further studies on tissue engineering. Imaging with expression of GFP facilitates the research on ASCs physiological behavior and application in tissue engineering during differentiation both in vitro and in vivo.
Collapse
Affiliation(s)
- Yunfeng Lin
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Lin Y, Luo E, Chen X, Liu L, Qiao J, Yan Z, Li Z, Tang W, Zheng X, Tian W. Molecular and cellular characterization during chondrogenic differentiation of adipose tissue-derived stromal cells in vitro and cartilage formation in vivo. J Cell Mol Med 2006; 9:929-39. [PMID: 16364200 PMCID: PMC6740126 DOI: 10.1111/j.1582-4934.2005.tb00389.x] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Human adipose tissue is a viable source of mesenchymal stem cells (MSCs) with wide differentiation potential for musculoskeletal tissue engineering research. The stem cell population, termed processed lipoaspirate (PLA) cells, can be isolated from human lipoaspirates and expanded in vitro easily. This study was to determine molecular and cellular characterization of PLA cells during chondrogenic differentiation in vitro and cartilage formation in vivo. When cultured in vitro with chondrogenic medium as monolayers in high density, they could be induced toward the chondrogenic lineages. To determine their ability of cartilage formation in vivo, the induced cells in alginate gel were implanted in nude mice subcutaneously for up to 20 weeks. Histological and immunohistochemical analysis of the induced cells and retrieved specimens from nude mice at various intervals showed obviously cartilaginous phenotype with positive staining of specific extracellular matrix (ECM). Correlatively, results of RT-PCR and Western Blot confirmed the expression of characteristic molecules during chondrogenic differentiation namely collagen type II, SOX9, cartilage oligomeric protein (COMP) and the cartilage-specific proteoglycan aggrecan. Meanwhile, there was low level synthesis of collagen type X and decreasing production of collagen type I during induction in vitro and formation of cartilaginous tissue in vivo. These cells induced to form engineered cartilage can maintain the stable phenotype and indicate no sign of hypertrophy in 20 weeks in vivo, however, when they cultured as monolayers, they showed prehypertrophic alteration in late stage about 10 weeks after induction. Therefore, it is suggested that human adipose tissue may represent a novel plentiful source of multipotential stem cells capable of undergoing chondrogenesis and forming engineered cartilage.
Collapse
Affiliation(s)
- Yunfeng Lin
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Lin Y, Chen X, Yan Z, Liu L, Tang W, Zheng X, Li Z, Qiao J, Li S, Tian W. Multilineage differentiation of adipose-derived stromal cells from GFP transgenic mice. Mol Cell Biochem 2006; 285:69-78. [PMID: 16477377 DOI: 10.1007/s11010-005-9056-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2005] [Accepted: 10/18/2005] [Indexed: 01/30/2023]
Abstract
Functional engineering of musculoskeletal tissues generally involves rapid expansion of progenitor cells in vitro while retaining their potential for further differentiation and then induction in specific culture conditions. The autologous adipose-derived stromal cells (ASCs) are considered to contain pluripotent mesenchymal stem cells. Imaging with expression of green fluorescent protein (GFP) facilitates the detailed research on ASCs physiological behavior during differentiation into a variety of cell lineages both in vitro and in vivo. In this study, we aimed to confirm the trans-germ plasticity of homogeneously marked ASCs from GFP transgenic mice. Simultaneously, the term and intensity of GFP expression in ASCs were also focused on during variant inductions, when cells were incubated with multiple growth factors and adjuvant. ASCs were harvested from inguinal fat pads of transgenic nude mice, passaged 3 times in monolayer cultures, and then transferred to osteogenic, adipogenic, neurogenic, and myogenic medium. The morphological characterization of inductive cells was observed using phase-contrast microscopy and histological staining such as alizarin red for mineralization nodules and oil red O for lipid accumulation. The expression of marker genes or proteins was measured using RT-PCR and immunocytochemical analysis. Collagen type I, osteopontin (OPN), and osteocalcin (OCN) were positive in osteogenic lineages, peroxisome proliferator-activated receptor(PPAR)-gamma2 and lipoprotein lipase (LPL) were positive in adipogenic ones, glial fibrillary acidic protein (GFAP) and neuron-specific enolase (NSE) were positive in neurogenic ones, and alpha-smooth muscle actin (alpha-SMA) was positive in myogenic ones. Moreover, the results of fluorescence microscopic imaging suggested that there was no significant decline of GFP expression during ASCs differentiation and the level of GFP maintained stable till differentiated ASCs showed apoptotic phenotype. So the endogenous GFP and multilineage potential of transgenic ASCs had no influences on each other. Since the population of GFP ASCs can be easily identified, it is proposed that they may be promising candidate seed cells for further studies on ASCs tissue engineering, especially the study on engineered tissues formed in vivo.
Collapse
Affiliation(s)
- Yunfeng Lin
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|