201
|
Harris Q, Seto J, O'Brien K, Lee PS, Kondo C, Heard BJ, Hart DA, Krawetz RJ. Monocyte chemotactic protein-1 inhibits chondrogenesis of synovial mesenchymal progenitor cells: an in vitro study. Stem Cells 2014; 31:2253-65. [PMID: 23836536 DOI: 10.1002/stem.1477] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 06/14/2013] [Indexed: 12/17/2022]
Abstract
Osteoarthritis (OA) is a multifactorial, often progressive, painful disease. OA often progresses with an apparent irreversible loss of articular cartilage, exposing underlying bone, resulting in pain and loss of mobility. This cartilage loss is thought to be permanent due to ineffective repair and apparent lack of stem/progenitor cells in that tissue. However, the adjacent synovial lining and synovial fluid are abundant with mesenchymal progenitor/stem cells (synovial mesenchymal progenitor cells [sMPCs]) capable of differentiating into cartilage both in vitro and in vivo. Previous studies have demonstrated that MPCs can home to factors such as monocyte chemotactic protein 1 (MCP-1/CCL2) expressed after injury. While MCP-1 (and its corresponding receptors) appears to play a role in recruiting stem cells to the site of injury, in this study, we have demonstrated that MCP-1 is upregulated in OA synovial fluid and that exposure to MCP-1 activates sMPCs, while concurrently inhibiting these cells from undergoing chondrogenesis in vitro. Furthermore, exposure to physiological (OA knee joint synovial fluid) levels of MCP-1 triggers changes in the transcriptome of sMPCs and prolonged exposure to the chemokine induces the expression of MCP-1 in sMPCs, resulting in a positive feedback loop from which sMPCs cannot apparently escape. Therefore, we propose a model where MCP-1 (normally expressed after joint injury) recruits sMPCs to the area of injury, but concurrently triggers changes in sMPC transcriptional regulation, leading to a blockage in the chondrogenic program. These results may open up new avenues of research into the lack of endogenous repair observed after articular cartilage injury and/or arthritis.
Collapse
Affiliation(s)
- Quinn Harris
- Department of Surgery, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | | | | | |
Collapse
|
202
|
Yu X, Khalil A, Dang PN, Alsberg E, Murphy WL. Multilayered Inorganic Microparticles for Tunable Dual Growth Factor Delivery. ADVANCED FUNCTIONAL MATERIALS 2014; 24:3082-3093. [PMID: 25342948 PMCID: PMC4204399 DOI: 10.1002/adfm.201302859] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
There is an increasing need to control the type, quantity, and timing of growth factors released during tissue healing. Sophisticated delivery systems offering the ability to deliver multiple growth factors with independently tunable kinetics are highly desirable. Here, a multilayered, mineral coated micro-particle (MCMs) platform that can serve as an adaptable dual growth factor delivery system is developed. Bone morphogenetic protein-2 (BMP-2) and vascular endothelial growth factor (VEGF) are bound to the mineral coatings with high binding efficiencies of up to 80%. BMP-2 is firstly bound onto a 1st mineral coating layer; then VEGF is bound onto a 2nd mineral coating layer. The release of BMP-2 is sustained over a period of 50 days while the release of VEGF is a typical two-phase release with rapid release in the first 14 days and more sustained release for the following 36 days. Notably, the release behaviors of both growth factors can be independently tailored by changing the intrinsic properties of the mineral coatings. Furthermore, the release of BMP-2 can be tuned by changing the thickness of the 2nd layer. This injectable microparticle based delivery platform with tunable growth factor release has immense potential for applications in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Xiaohua Yu
- Department of Biomedical Engineering, University of Wisconsin, 1111 Highland Ave, Madison, WI, 53705, USA
| | - Andrew Khalil
- Department of Biomedical Engineering, University of Wisconsin, 1111 Highland Ave, Madison, WI, 53705, USA
| | - Phuong Ngoc Dang
- Department of Biomedical Engineering and Orthopaedic Surgery, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Eben Alsberg
- Department of Biomedical Engineering and Orthopaedic Surgery, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
- AO Foundation Collaborative Research Center, Clavadelerstrasse 8, Davos, 7270, Switzerland
| | - William L. Murphy
- Department of Biomedical Engineering, University of Wisconsin, 1111 Highland Ave, Madison, WI, 53705, USA
- AO Foundation Collaborative Research Center, Clavadelerstrasse 8, Davos, 7270, Switzerland
- Department of Orthopedics and Rehabilitation, 1300 University Ave, University of Wisconsin, Madison, WI, 53705, USA
| |
Collapse
|
203
|
Lei M, Li K, Li B, Gao LN, Chen FM, Jin Y. Mesenchymal stem cell characteristics of dental pulp and periodontal ligament stem cells after in vivo transplantation. Biomaterials 2014; 35:6332-43. [PMID: 24824581 DOI: 10.1016/j.biomaterials.2014.04.071] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 04/17/2014] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) isolated from human postnatal dental pulp and periodontal ligament (PDL) tissues can give rise to multilineage differentiation in vitro and generate related dental tissues in vivo. However, the cell properties of human dental pulp stem cells (DPSCs) and PDL stem cells (PDLSCs) after in vivo implantation remain largely unidentified. In this study, cells were re-isolated from in vivo-generated dental pulp-like and PDL-like tissues (termed re-DPCs and re-PDLCs, respectively) as a result of ectopic transplantation of human DPSC and PDLSC sheets. The cell characteristics in terms of colony-forming ability, cell surface antigens and multi-differentiation potentials were all evaluated before and after implantation. It was found that re-DPCs and re-PDLCs were of human and mesenchymal origin and positive for MSC markers such as STRO-1, CD146, CD29, CD90 and CD105; and, to some extent, re-DPCs could maintain their colony forming abilities. Moreover, both cell types were able to form mineral deposits and differentiate into adipocytes and chondrocytes; however, quantitative analysis and related gene expression determination showed that the osteo-/chondro-differentiation capabilities of re-DPCs and re-PDLCs were significantly reduced compared to those of DPSCs and PDLSCs, respectively (P < 0.05); re-PDLCs showed a greater reduction potential than re-DPCs. We conclude that DPSCs and PDLSCs may maintain their MSC characteristics after in vivo implantation and, compared to PDLSCs, DPSCs appear much more stable under in vivo conditions. These findings provide additional cellular and molecular evidence that supports expanding the use of dental tissue-derived stem cells in cell therapy and tissue engineering.
Collapse
Affiliation(s)
- Ming Lei
- State Key Laboratory of Military Stomatology, Department of Periodontology & Biomaterials Unit, School of Stomatology, Fourth Military Medical University, Xi'an, PR China; State Key Laboratory of Military Stomatology, Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, PR China
| | - Kun Li
- State Key Laboratory of Military Stomatology, Department of Periodontology & Biomaterials Unit, School of Stomatology, Fourth Military Medical University, Xi'an, PR China; State Key Laboratory of Military Stomatology, Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, PR China
| | - Bei Li
- State Key Laboratory of Military Stomatology, Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, PR China
| | - Li-Na Gao
- State Key Laboratory of Military Stomatology, Department of Periodontology & Biomaterials Unit, School of Stomatology, Fourth Military Medical University, Xi'an, PR China; State Key Laboratory of Military Stomatology, Translational Research Team, School of Stomatology, Fourth Military Medical University, Xi'an, PR China
| | - Fa-Ming Chen
- State Key Laboratory of Military Stomatology, Department of Periodontology & Biomaterials Unit, School of Stomatology, Fourth Military Medical University, Xi'an, PR China; State Key Laboratory of Military Stomatology, Translational Research Team, School of Stomatology, Fourth Military Medical University, Xi'an, PR China.
| | - Yan Jin
- State Key Laboratory of Military Stomatology, Research and Development Center for Tissue Engineering, Fourth Military Medical University, Xi'an, PR China; State Key Laboratory of Military Stomatology, Translational Research Team, School of Stomatology, Fourth Military Medical University, Xi'an, PR China.
| |
Collapse
|
204
|
Pan S, Dangaria S, Gopinathan G, Yan X, Lu X, Kolokythas A, Niu Y, Luan X. SCF promotes dental pulp progenitor migration, neovascularization, and collagen remodeling - potential applications as a homing factor in dental pulp regeneration. Stem Cell Rev Rep 2014; 9:655-67. [PMID: 23703692 DOI: 10.1007/s12015-013-9442-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Stem cell factor (SCF) is a powerful chemokine that binds to the c-Kit receptor CD117 and has shown promise as a homing agent capable of progenitor cell recruitment. In the present study we have documented high levels of both SCF and its receptor c-Kit in differentiating dental pulp (DP) cells and in the sub-odontoblastic layer of Höhl. In vitro studies using human DP progenitors revealed a significant increase in cell proliferation after100 nM SCF application, explained by a 2-fold upregulation in cyclin D3 and FGF2 cell cycle regulators, and a 7-fold increase in CDK4 expression. DP cell migration in the presence of SCF was up-regulated 2.7-fold after a 24 h culture period, and this effect was accompanied by cytoskeletal rearrangement, a 1.5-fold increase in polymeric F-actin over G-actin, and a 1.8-fold increase in RhoA expression. Explaining the signaling effect of SCF on DP migration, PI3K/Akt and MEK/ERK pathway inhibitors were demonstrated to significantly reduce DP cell migration, while SCF alone doubled the number of migrated cells. ERK and AKT phosphorylation were dramatically upregulated already 3-5 min after SCF addition to the culture medium and declined thereafter, classifying SCF as a fast acting chemokine. When applied as an agent to promote tissue regeneration in subcutaneously implanted collagen sponges, SCF resulted in a 7-fold increase in the cell number in the implanted tissue construct, a more than 9-fold increase in capillaries, as well as collagen sponge remodeling and collagen fiber neogenesis. Together, these studies demonstrate the suitability of SCF as a potent aid in the regeneration of dental pulp and other mesenchymal tissues, capable of inducing cell homing, angiogenesis, and tissue remodeling.
Collapse
Affiliation(s)
- Shuang Pan
- School of Dentistry, Department of Endodontics, Harbin Medical University, Harbin, China
| | | | | | | | | | | | | | | |
Collapse
|
205
|
Monsarrat P, Vergnes JN, Nabet C, Sixou M, Snead ML, Planat-Bénard V, Casteilla L, Kémoun P. Concise review: mesenchymal stromal cells used for periodontal regeneration: a systematic review. Stem Cells Transl Med 2014; 3:768-74. [PMID: 24744392 DOI: 10.5966/sctm.2013-0183] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Periodontitis is a chronic infectious disease of the soft and hard tissues supporting the teeth. Recent advances in regenerative medicine and stem cell biology have paved the way for periodontal tissue engineering. Mesenchymal stromal cells (MSCs) delivered in situ to periodontal defects may exert their effects at multiple levels, including neovascularization, immunomodulation, and tissue regeneration. This systematic review had two goals: (a) to objectively quantify key elements for efficacy and safety of MSCs used for periodontal regeneration and (b) to identify patterns in the existing literature to explain differences between studies and suggest recommendations for future research. This systematic review provided good evidence of the capacity of MSCs to regenerate periodontal tissues in animals; however, experimentally generated defects used in animal studies do not sufficiently mimic the pathophysiology of periodontitis in humans. Moreover, the safety of such interventions in humans still needs to be studied. There were marked differences between experimental and control groups that may be influenced by characteristics that are crucial to address before translation to human clinical trials. We suggest that the appropriate combination of cell source, carrier type, and biomolecules, as well as the inclusion of critical path issues for a given clinical case, should be further explored and refined before transitioning to clinical trials. Future studies should investigate periodontal regenerative procedures in animal models, including rodents, in which the defects generated are designed to more accurately reflect the inflammatory status of the host and the shift in their pathogenic microflora.
Collapse
Affiliation(s)
- Paul Monsarrat
- STROMALab, Toulouse, France; Université de Toulouse, Toulouse, France; INSERM, Toulouse, France; Etablissement Français du Sang Pyrénées-Méditerranée, Toulouse, France; Departments of Public Health and Biology, Toulouse Faculty of Dentistry, Paul Sabatier University and Toulouse University Hospital, CHU de Toulouse, Toulouse, France; Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, USA
| | - Jean-Noël Vergnes
- STROMALab, Toulouse, France; Université de Toulouse, Toulouse, France; INSERM, Toulouse, France; Etablissement Français du Sang Pyrénées-Méditerranée, Toulouse, France; Departments of Public Health and Biology, Toulouse Faculty of Dentistry, Paul Sabatier University and Toulouse University Hospital, CHU de Toulouse, Toulouse, France; Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, USA
| | - Cathy Nabet
- STROMALab, Toulouse, France; Université de Toulouse, Toulouse, France; INSERM, Toulouse, France; Etablissement Français du Sang Pyrénées-Méditerranée, Toulouse, France; Departments of Public Health and Biology, Toulouse Faculty of Dentistry, Paul Sabatier University and Toulouse University Hospital, CHU de Toulouse, Toulouse, France; Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, USA
| | - Michel Sixou
- STROMALab, Toulouse, France; Université de Toulouse, Toulouse, France; INSERM, Toulouse, France; Etablissement Français du Sang Pyrénées-Méditerranée, Toulouse, France; Departments of Public Health and Biology, Toulouse Faculty of Dentistry, Paul Sabatier University and Toulouse University Hospital, CHU de Toulouse, Toulouse, France; Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, USA
| | - Malcolm L Snead
- STROMALab, Toulouse, France; Université de Toulouse, Toulouse, France; INSERM, Toulouse, France; Etablissement Français du Sang Pyrénées-Méditerranée, Toulouse, France; Departments of Public Health and Biology, Toulouse Faculty of Dentistry, Paul Sabatier University and Toulouse University Hospital, CHU de Toulouse, Toulouse, France; Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, USA
| | - Valérie Planat-Bénard
- STROMALab, Toulouse, France; Université de Toulouse, Toulouse, France; INSERM, Toulouse, France; Etablissement Français du Sang Pyrénées-Méditerranée, Toulouse, France; Departments of Public Health and Biology, Toulouse Faculty of Dentistry, Paul Sabatier University and Toulouse University Hospital, CHU de Toulouse, Toulouse, France; Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, USA
| | - Louis Casteilla
- STROMALab, Toulouse, France; Université de Toulouse, Toulouse, France; INSERM, Toulouse, France; Etablissement Français du Sang Pyrénées-Méditerranée, Toulouse, France; Departments of Public Health and Biology, Toulouse Faculty of Dentistry, Paul Sabatier University and Toulouse University Hospital, CHU de Toulouse, Toulouse, France; Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, USA
| | - Philippe Kémoun
- STROMALab, Toulouse, France; Université de Toulouse, Toulouse, France; INSERM, Toulouse, France; Etablissement Français du Sang Pyrénées-Méditerranée, Toulouse, France; Departments of Public Health and Biology, Toulouse Faculty of Dentistry, Paul Sabatier University and Toulouse University Hospital, CHU de Toulouse, Toulouse, France; Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
206
|
Shao Z, Zhang X, Pi Y, Yin L, Li L, Chen H, Zhou C, Ao Y. Surface modification on polycaprolactone electrospun mesh and human decalcified bone scaffold with synovium-derived mesenchymal stem cells-affinity peptide for tissue engineering. J Biomed Mater Res A 2014; 103:318-29. [PMID: 24659568 DOI: 10.1002/jbm.a.35177] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 03/11/2014] [Accepted: 03/21/2014] [Indexed: 12/13/2022]
Abstract
Synovium-derived mesenchymal stem cells (SMSC) have been studied for over a decade since first being successfully isolated in 2001. These cells demonstrate the most promising therapeutic efficacy for musculoskeletal regeneration of the MSC family, particularly for cartilage regeneration. However, the mobilization and transfer of MSCs to defective or damaged tissues and organs in vivo with high accuracy and efficiency has been a major problem in tissue engineering (TE). In the present study, we identified a seven amino acid peptide sequence [SMSCs-affinity peptide (LTHPRWP; L7)] through phage display technology that has a high specific affinity to SMSCs. Our analysis suggested that L7 efficiently and specifically interacted with SMSCs without any species specificity. Thereafter, L7 was covalently conjugated onto both polycaprolactone (PCL) electrospun meshes and human decalcified bone scaffolds (hDBSc) to investigate its TE applications. After 24 h coculture with human SMSCs (hSMSCs), L7-conjugated PCL electrospun meshes had significantly more adherent hSMSCs than the control group, and the cells expanded well. Similar results were obtained using hDBSs. These results suggest that the novel L7 peptide sequence has a high specific affinity to SMSCs. Covalently conjugating this peptide to either artificial polymer material (PCL mesh) or natural material (hDBS) significantly enhances the adhesion of SMSCs. This method is applicable to a wide range of potential SMSC-based TE applications, particularly to cartilage regeneration, via surface modification on various type of materials.
Collapse
Affiliation(s)
- Zhenxing Shao
- Institute of Sports Medicine, Peking University Third Hospital, Haidian District, Beijing, 100191, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
207
|
Cartilage tissue engineering: molecular control of chondrocyte differentiation for proper cartilage matrix reconstruction. Biochim Biophys Acta Gen Subj 2014; 1840:2414-40. [PMID: 24608030 DOI: 10.1016/j.bbagen.2014.02.030] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 02/06/2014] [Accepted: 02/26/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND Articular cartilage defects are a veritable therapeutic problem because therapeutic options are very scarce. Due to the poor self-regeneration capacity of cartilage, minor cartilage defects often lead to osteoarthritis. Several surgical strategies have been developed to repair damaged cartilage. Autologous chondrocyte implantation (ACI) gives encouraging results, but this cell-based therapy involves a step of chondrocyte expansion in a monolayer, which results in the loss in the differentiated phenotype. Thus, despite improvement in the quality of life for patients, reconstructed cartilage is in fact fibrocartilage. Successful ACI, according to the particular physiology of chondrocytes in vitro, requires active and phenotypically stabilized chondrocytes. SCOPE OF REVIEW This review describes the unique physiology of cartilage, with the factors involved in its formation, stabilization and degradation. Then, we focus on some of the most recent advances in cell therapy and tissue engineering that open up interesting perspectives for maintaining or obtaining the chondrogenic character of cells in order to treat cartilage lesions. MAJOR CONCLUSIONS Current research involves the use of chondrocytes or progenitor stem cells, associated with "smart" biomaterials and growth factors. Other influential factors, such as cell sources, oxygen pressure and mechanical strain are considered, as are recent developments in gene therapy to control the chondrocyte differentiation/dedifferentiation process. GENERAL SIGNIFICANCE This review provides new information on the mechanisms regulating the state of differentiation of chondrocytes and the chondrogenesis of mesenchymal stem cells that will lead to the development of new restorative cell therapy approaches in humans. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.
Collapse
|
208
|
Gattazzo F, Urciuolo A, Bonaldo P. Extracellular matrix: a dynamic microenvironment for stem cell niche. Biochim Biophys Acta Gen Subj 2014; 1840:2506-19. [PMID: 24418517 PMCID: PMC4081568 DOI: 10.1016/j.bbagen.2014.01.010] [Citation(s) in RCA: 849] [Impact Index Per Article: 84.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 01/05/2014] [Accepted: 01/06/2014] [Indexed: 02/08/2023]
Abstract
Background Extracellular matrix (ECM) is a dynamic and complex environment characterized by biophysical, mechanical and biochemical properties specific for each tissue and able to regulate cell behavior. Stem cells have a key role in the maintenance and regeneration of tissues and they are located in a specific microenvironment, defined as niche. Scope of review We overview the progresses that have been made in elucidating stem cell niches and discuss the mechanisms by which ECM affects stem cell behavior. We also summarize the current tools and experimental models for studying ECM–stem cell interactions. Major conclusions ECM represents an essential player in stem cell niche, since it can directly or indirectly modulate the maintenance, proliferation, self-renewal and differentiation of stem cells. Several ECM molecules play regulatory functions for different types of stem cells, and based on its molecular composition the ECM can be deposited and finely tuned for providing the most appropriate niche for stem cells in the various tissues. Engineered biomaterials able to mimic the in vivo characteristics of stem cell niche provide suitable in vitro tools for dissecting the different roles exerted by the ECM and its molecular components on stem cell behavior. General significance ECM is a key component of stem cell niches and is involved in various aspects of stem cell behavior, thus having a major impact on tissue homeostasis and regeneration under physiological and pathological conditions. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties. Stem cells have a key role in the maintenance and regeneration of tissues. The extracellular matrix is a critical regulator of stem cell function. Stem cells reside in a dynamic and specialized microenvironment denoted as niche. The extracellular matrix represents an essential component of stem cell niches. Bioengineered niches can be used for investigating stem cell–matrix interactions.
Collapse
Affiliation(s)
- Francesca Gattazzo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Anna Urciuolo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy.
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy.
| |
Collapse
|
209
|
Elisseeff J, Madrid MG, Lu Q, Chae JJ, Guo Q. Future perspectives for regenerative medicine in ophthalmology. Middle East Afr J Ophthalmol 2014; 20:38-45. [PMID: 23580850 PMCID: PMC3617526 DOI: 10.4103/0974-9233.106385] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Repair and reconstruction of the cornea has historically relied on synthetic materials or tissue transplantation. However, the future holds promise for treatments using smart biomaterials and stem cells that direct tissue repair and regeneration to ultimately create new ocular structures that are indistinguishable from the original native tissue. The cornea is a remarkable engineering structure. By understanding the physical structure of the tissue and the resulting impact of the structure on biological function, we can design novel materials for a number of ophthalmic clinical applications. Furthermore, by extending this structure-function approach to characterizing corneal disease processes, new therapies can be engineered.
Collapse
Affiliation(s)
- Jennifer Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | | | | | | |
Collapse
|
210
|
A review of stem cell translation and potential confounds by cancer stem cells. Stem Cells Int 2013; 2013:241048. [PMID: 24385986 PMCID: PMC3872439 DOI: 10.1155/2013/241048] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 11/05/2013] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells found in both fetal and adult tissues. MSCs show promise for cellular therapy for several disorders such as those associated with inflammation. In adults, MSCs primarily reside in the bone marrow (BM) and adipose tissues. In BM, MSCs are found at low frequency around blood vessels and trabecula. MSCs are attractive candidates for regenerative medicine given their ease in harvesting and expansion and their unique ability to bypass the immune system in an allogeneic host. Additionally, MSCs exert pathotropism by their ability to migrate to diseased regions. Despite the "attractive" properties of MSCs, their translation to patients requires indepth research. "Off-the-shelf" MSCs are proposed for use in an allogeneic host. Thus, the transplanted MSCs, when placed in a foreign host, could receive cue from the microenvironment for cellular transformation. An important problem with the use of MSCs involves their ability to facilitate the support of breast and other cancers as carcinoma-associated fibroblasts. MSCs could show distinct effect on each subset of cancer cells. This could lead to untoward effect during MSC therapy since the MSCs would be able to interact with undiagnosed cancer cells, which might be in a dormant state. Based on these arguments, further preclinical research is needed to ensure patient safety with MSC therapy. Here, we discuss the basic biology of MSCs, discuss current applications, and provide evidence why it is important to understand MSC biology in the context of diseased microenvironment for safe application.
Collapse
|
211
|
Enhancing the migration ability of mesenchymal stromal cells by targeting the SDF-1/CXCR4 axis. BIOMED RESEARCH INTERNATIONAL 2013; 2013:561098. [PMID: 24381939 PMCID: PMC3870125 DOI: 10.1155/2013/561098] [Citation(s) in RCA: 200] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 10/09/2013] [Accepted: 10/28/2013] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal cells (MSCs) are currently being investigated in numerous clinical trials of tissue repair and various immunological disorders based on their ability to secrete trophic factors and to modulate inflammatory responses. MSCs have been shown to migrate to sites of injury and inflammation in response to soluble mediators including the chemokine stromal cell-derived factor-(SDF-)1, but during in vitro culture expansion MSCs lose surface expression of key homing receptors particularly of the SDF-1 receptor, CXCR4. Here we review studies on enhancement of SDF-1-directed migration of MSCs with the premise that their improved recruitment could translate to therapeutic benefits. We describe our studies on approaches to increase the CXCR4 expression in in vitro-expanded cord blood-derived MSCs, namely, transfection, using the commercial liposomal reagent IBAfect, chemical treatment with the histone deacetylase inhibitor valproic acid, and exposure to recombinant complement component C1q. These methodologies will be presented in the context of other cell targeting and delivery strategies that exploit pathways involved in MSC migration. Taken together, these findings indicate that MSCs can be manipulated in vitro to enhance their in vivo recruitment and efficacy for tissue repair.
Collapse
|
212
|
Trofin EA, Monsarrat P, Kémoun P. Cell therapy of periodontium: from animal to human? Front Physiol 2013; 4:325. [PMID: 24298258 PMCID: PMC3828527 DOI: 10.3389/fphys.2013.00325] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 10/19/2013] [Indexed: 12/30/2022] Open
Abstract
Periodontitis is a chronic inflammatory disease affecting the soft and hard tissues supporting the teeth, which often leads to tooth loss. Its significant impact on the patient's general health and quality of life point to a need for more effective management of this condition. Existing treatments include scaling/root planning and surgical approaches but their overall effects are relatively modest and restricted in application. The goal of regenerative therapy of periodontal defects is to enhance endogenous progenitors and thus promote optimal wound healing. Considering that the host or tissue might be defective in the periodontitis context, it has been proposed that grafting exogenous stem cells would produce new tissues and create a suitable microenvironment for tissue regeneration. Thus, cell therapy of periodontium has been assessed in many animal models and promising results have been reported. However, the methodological diversity of these studies makes the conversion to clinical practice difficult. The aim of this review is to highlight the primary requirements to be satisfied before the leap to clinical trials can be made. We therefore review cell therapy applications for periodontal regeneration in animal models and the concerns to be addressed before undertaking human experiments.
Collapse
Affiliation(s)
- Elena A Trofin
- Department of Biology, Toulouse Faculty of Dentistry - Paul Sabatier University, Toulouse University Hospital - CHU de Toulouse Toulouse, France ; Department of Pathology, Faculty of Medicine and Odontology, University of Valencia Valencia, Spain
| | | | | |
Collapse
|
213
|
Teixeira FG, Carvalho MM, Sousa N, Salgado AJ. Mesenchymal stem cells secretome: a new paradigm for central nervous system regeneration? Cell Mol Life Sci 2013; 70:3871-82. [PMID: 23456256 PMCID: PMC11113366 DOI: 10.1007/s00018-013-1290-8] [Citation(s) in RCA: 220] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 01/22/2013] [Accepted: 02/04/2013] [Indexed: 12/15/2022]
Abstract
The low regeneration potential of the central nervous system (CNS) represents a challenge for the development of new therapeutic strategies. Mesenchymal stem cells (MSCs) have been proposed as a possible therapeutic tool for CNS disorders. In addition to their differentiation potential, it is well accepted nowadays that their beneficial actions can also be mediated by their secretome. Indeed, it was already demonstrated, both in vitro and in vivo, that MSCs are able to secrete a broad range of neuroregulatory factors that promote an increase in neurogenesis, inhibition of apoptosis and glial scar formation, immunomodulation, angiogenesis, neuronal and glial cell survival, as well as relevant neuroprotective actions on different pathophysiological contexts. Considering their protective action in lesioned sites, MSCs' secretome might also improve the integration of local progenitor cells in neuroregeneration processes, opening a door for their future use as therapeutical strategies in human clinical trials. Thus, in this review we analyze the current understanding of MSCs secretome as a new paradigm for the treatment of CNS neurodegenerative diseases.
Collapse
Affiliation(s)
- Fábio G. Teixeira
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Miguel M. Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - António J. Salgado
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
214
|
Toeg HD, Tiwari-Pandey R, Seymour R, Ahmadi A, Crowe S, Vulesevic B, Suuronen EJ, Ruel M. Injectable small intestine submucosal extracellular matrix in an acute myocardial infarction model. Ann Thorac Surg 2013; 96:1686-94; discussion 1694. [PMID: 24083799 DOI: 10.1016/j.athoracsur.2013.06.063] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 06/03/2013] [Accepted: 06/06/2013] [Indexed: 12/24/2022]
Abstract
BACKGROUND The mechanisms involved in myocardial regeneration and cardiac remodeling were examined by injecting porcine-derived small intestine submucosal extracellular matrix (SIS-ECM), with and without circulating angiogenic cells (CACs), in a mouse model of acute myocardial infarction (MI). METHODS Nine- to 10-week-old female C57BL/6J mice had the left anterior descending (LAD) coronary artery ligated. Seven days after ligation, 38 randomly allocated animals received echocardiographically guided intramyocardial injections of phosphate buffered saline (PBS), CACs, SIS-ECM, or SIS-ECM + CACs. Repeated echocardiography and immunohistochemical analysis were performed at 28 days after ligation. RESULTS Baseline postligation left ventricular ejection fraction (LVEF) was equivalent in all groups. Twenty-one days after treatment, ejection fraction improved in the SIS-ECM + CAC treatment group (by 38% ± 2.12%) and the SIS-ECM treatment group (by 36% ± 3.71%), compared with the CAC-alone and PBS treatment groups (p < 0.1). Masson's trichrome staining showed reduced infarct size in SIS-ECM + CACs (34.2% ± 3.1%) and SIS-ECM alone (34.5% ± 4.7%) compared with CACs alone (47.3% ± 6.0%) and PBS (61.9% ± 5.5%; p < 0.002). Arteriolar density in periinfarct regions was enhanced in both SIS-ECM-treated groups (by ≥ 78% ± 7%; p = 0.03). More GATA4- and β-catenin-positive cardiac cells were found in the myocardium of SIS-ECM-treated animals. CONCLUSIONS Intramyocardial delivery of SIS-ECM 7 days after MI in a mouse model reduced infarct size and improved myocardial vessel density and function; when combined with CACs it helped restore myocardial cellularity, suggesting a potential therapeutic role for SIS-ECM in cardiac regeneration.
Collapse
Affiliation(s)
- Hadi Daood Toeg
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
215
|
English D, Sharma NK, Sharma K, Anand A. Neural stem cells-trends and advances. J Cell Biochem 2013; 114:764-72. [PMID: 23225161 DOI: 10.1002/jcb.24436] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 10/23/2012] [Indexed: 12/12/2022]
Abstract
For many years, accepted dogma held that brain is a static organ with no possibility of regeneration of cells in injured or diseased human brain. However, recent preclinical reports have shown regenerative potential of neural stem cells using various injury models. This has resulted in renewed hope for those suffering from spinal cord injury and neural damage. As the potential of stem cell therapy gained impact, these claims, in particular, led to widespread enthusiasm that acute and chronic injury of the nervous system would soon be a problem of the past. The devastation caused by injury or diseases of the brain and spinal cord led to wide premature acceptance that "neural stem cells (NSCs)" derived from embryonic, fetal or adult sources would soon be effective in reversing neural and spinal trauma. However, neural therapy with stem cells has not been realized to its fullest extent. Although, discrete population of regenerative stem cells seems to be present in specific areas of human brain, the function of these cells is unclear. However, similar cells in animals seem to play important role in postnatal growth as well as recovery of neural tissue from injury, anoxia, or disease.
Collapse
Affiliation(s)
- Denis English
- Foundation for Florida Development and Research, Palmetto, Florida
| | | | | | | |
Collapse
|
216
|
Yang H, Gao LN, An Y, Hu CH, Jin F, Zhou J, Jin Y, Chen FM. Comparison of mesenchymal stem cells derived from gingival tissue and periodontal ligament in different incubation conditions. Biomaterials 2013; 34:7033-47. [DOI: 10.1016/j.biomaterials.2013.05.025] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Accepted: 05/16/2013] [Indexed: 12/25/2022]
|
217
|
Zhang K, Liu T, Li JA, Chen JY, Wang J, Huang N. Surface modification of implanted cardiovascular metal stents: From antithrombosis and antirestenosis to endothelialization. J Biomed Mater Res A 2013; 102:588-609. [PMID: 23520056 DOI: 10.1002/jbm.a.34714] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Revised: 03/08/2013] [Accepted: 03/11/2013] [Indexed: 12/31/2022]
Affiliation(s)
- Kun Zhang
- Key Laboratory of Advanced Technology for Materials of Chinese Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, People's Republic of China
| | | | | | | | | | | |
Collapse
|
218
|
Yuan JM, Xiong SH, Liu Z, Wen Y, Dang RS, Shen MR, Zhang YZ, Zhang X, Yang XQ, Zhang CS. Functional analysisin vivoof engineered valved venous conduit with decellularized matrix and two bone marrow-derived progenitors in sheep. J Tissue Eng Regen Med 2013; 10:554-63. [DOI: 10.1002/term.1748] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 10/24/2012] [Accepted: 03/14/2013] [Indexed: 11/10/2022]
Affiliation(s)
- Jian-Ming Yuan
- Department of Anatomy; Second Military Medical University; Shanghai People's Republic of China
| | - Shao-Hu Xiong
- Department of Anatomy; Second Military Medical University; Shanghai People's Republic of China
| | - Zhen Liu
- Department of Anatomy; Second Military Medical University; Shanghai People's Republic of China
| | - Yu Wen
- Department of Anatomy; Second Military Medical University; Shanghai People's Republic of China
| | - Rui-Shan Dang
- Department of Anatomy; Second Military Medical University; Shanghai People's Republic of China
| | - Man-Ru Shen
- Department of Anatomy; Second Military Medical University; Shanghai People's Republic of China
| | - Yong-Zhen Zhang
- Department of Anatomy; Second Military Medical University; Shanghai People's Republic of China
| | - Xi Zhang
- Department of Anatomy; Second Military Medical University; Shanghai People's Republic of China
| | - Xiang-Qun Yang
- Department of Anatomy; Second Military Medical University; Shanghai People's Republic of China
| | - Chuan-Sen Zhang
- Department of Anatomy; Second Military Medical University; Shanghai People's Republic of China
| |
Collapse
|
219
|
Vanden Berg-Foels WS. In situ tissue regeneration: chemoattractants for endogenous stem cell recruitment. TISSUE ENGINEERING PART B-REVIEWS 2013; 20:28-39. [PMID: 23678952 DOI: 10.1089/ten.teb.2013.0100] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tissue engineering uses cells, signaling molecules, and/or biomaterials to regenerate injured or diseased tissues. Ex vivo expanded mesenchymal stem cells (MSC) have long been a cornerstone of regeneration therapies; however, drawbacks that include altered signaling responses and reduced homing capacity have prompted investigation of regeneration based on endogenous MSC recruitment. Recent successful proof-of-concept studies have further motivated endogenous MSC recruitment-based approaches. Stem cell migration is required for morphogenesis and organogenesis during development and for tissue maintenance and injury repair in adults. A biomimetic approach to in situ tissue regeneration by endogenous MSC requires the orchestration of three main stages: MSC recruitment, MSC differentiation, and neotissue maturation. The first stage must result in recruitment of a sufficient number of MSC, capable of effecting regeneration, to the injured or diseased tissue. One of the challenges for engineering endogenous MSC recruitment is the selection of effective chemoattractant(s). The objective of this review is to synthesize and evaluate evidence of recruitment efficacy by reported chemoattractants, including growth factors, chemokines, and other more recently appreciated MSC chemoattractants. The influence of MSC tissue sources, cell culture methods, and the in vitro and in vivo environments is discussed. This growing body of knowledge will serve as a basis for the rational design of regenerative therapies based on endogenous MSC recruitment. Successful endogenous MSC recruitment is the first step of successful tissue regeneration.
Collapse
|
220
|
The combined use of cell sheet fragments of periodontal ligament stem cells and platelet-rich fibrin granules for avulsed tooth reimplantation. Biomaterials 2013; 34:5506-20. [DOI: 10.1016/j.biomaterials.2013.03.079] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 03/27/2013] [Indexed: 01/27/2023]
|
221
|
Mirabella T, Gentili C, Daga A, Cancedda R. Amniotic fluid stem cells in a bone microenvironment: Driving host angiogenic response. Stem Cell Res 2013; 11:540-51. [DOI: 10.1016/j.scr.2013.02.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 02/10/2013] [Accepted: 02/11/2013] [Indexed: 12/17/2022] Open
|
222
|
Chen FM, Lu H, Wu LA, Gao LN, An Y, Zhang J. Surface-engineering of glycidyl methacrylated dextran/gelatin microcapsules with thermo-responsive poly(N-isopropylacrylamide) gates for controlled delivery of stromal cell-derived factor-1α. Biomaterials 2013; 34:6515-27. [PMID: 23726519 DOI: 10.1016/j.biomaterials.2013.05.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 05/07/2013] [Indexed: 12/18/2022]
Abstract
In situ tissue engineering has been proposed as a promising method to address the need for the clinical regeneration of a wide variety of damaged tissues. This approach comprises the use of a cell-free instructive scaffold that incorporates and releases topical chemotactic factors to recruit host endogenous stem/progenitor cells for tissue regrowth at the locus of implantation. However, the clinical translation of this concept is hampered when repeated doses of medication must be administrated over an extended period of time. In this study, we designed a delivery platform characterized by microcapsules containing thermo-responsive poly(N-isopropylacrylamide) (PNIPAAm) gates on their outer pore surfaces for the controlled release of stromal cell-derived factor (SDF)-1α, an important chemokine for stem cell recruitment/homing. Double-phase emulsified condensation polymerization was used to prepare interconnected porous glycidyl methacrylated dextran (Dex-GMA)/gelatin microcapsules, and plasma-graft pore-filling polymerization was used to graft PNIPAAm into the surface pores of the microcapsules. The in vitro results showed that the PNIPAAm-grafted microcapsules featured thermo-responsive drug release properties due to the swollen-shrunken property of PNIPAAm gates in response to temperature changes. After subcutaneous implantation, the thermally responsive microcapsules resulted in a more sustained and long-term SDF-1α release compared with those without PNIPAAm-grafting. In the future, this delivery system may have great potential for use in cell recruiting biomaterials for various tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Fa-Ming Chen
- Department of Periodontology and Oral Medicine, School of Stomatology, Fourth Military Medical University, Xi'an 710032, Shaanxi, PR China.
| | | | | | | | | | | |
Collapse
|
223
|
Amini AR, Laurencin CT, Nukavarapu SP. Bone tissue engineering: recent advances and challenges. Crit Rev Biomed Eng 2013; 40:363-408. [PMID: 23339648 DOI: 10.1615/critrevbiomedeng.v40.i5.10] [Citation(s) in RCA: 1350] [Impact Index Per Article: 122.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The worldwide incidence of bone disorders and conditions has trended steeply upward and is expected to double by 2020, especially in populations where aging is coupled with increased obesity and poor physical activity. Engineered bone tissue has been viewed as a potential alternative to the conventional use of bone grafts, due to their limitless supply and no disease transmission. However, bone tissue engineering practices have not proceeded to clinical practice due to several limitations or challenges. Bone tissue engineering aims to induce new functional bone regeneration via the synergistic combination of biomaterials, cells, and factor therapy. In this review, we discuss the fundamentals of bone tissue engineering, highlighting the current state of this field. Further, we review the recent advances of biomaterial and cell-based research, as well as approaches used to enhance bone regeneration. Specifically, we discuss widely investigated biomaterial scaffolds, micro- and nano-structural properties of these scaffolds, and the incorporation of biomimetic properties and/or growth factors. In addition, we examine various cellular approaches, including the use of mesenchymal stem cells (MSCs), embryonic stem cells (ESCs), adult stem cells, induced pluripotent stem cells (iPSCs), and platelet-rich plasma (PRP), and their clinical application strengths and limitations. We conclude by overviewing the challenges that face the bone tissue engineering field, such as the lack of sufficient vascularization at the defect site, and the research aimed at functional bone tissue engineering. These challenges will drive future research in the field.
Collapse
Affiliation(s)
- Ami R Amini
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, CT, USA
| | | | | |
Collapse
|
224
|
Rennert RC, Sorkin M, Garg RK, Gurtner GC. Stem cell recruitment after injury: lessons for regenerative medicine. Regen Med 2013; 7:833-50. [PMID: 23164083 DOI: 10.2217/rme.12.82] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tissue repair and regeneration are thought to involve resident cell proliferation as well as the selective recruitment of circulating stem and progenitor cell populations through complex signaling cascades. Many of these recruited cells originate from the bone marrow, and specific subpopulations of bone marrow cells have been isolated and used to augment adult tissue regeneration in preclinical models. Clinical studies of cell-based therapies have reported mixed results, however, and a variety of approaches to enhance the regenerative capacity of stem cell therapies are being developed based on emerging insights into the mechanisms of progenitor cell biology and recruitment following injury. This article discusses the function and mechanisms of recruitment of important bone marrow-derived stem and progenitor cell populations following injury, as well as the emerging therapeutic applications targeting these cells.
Collapse
Affiliation(s)
- Robert C Rennert
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, 257 Campus Drive West, Hagey Building GK-201, Stanford, CA 94305-5148, USA
| | | | | | | |
Collapse
|
225
|
Kyburz KA, Anseth KS. Three-dimensional hMSC motility within peptide-functionalized PEG-based hydrogels of varying adhesivity and crosslinking density. Acta Biomater 2013; 9:6381-92. [PMID: 23376239 DOI: 10.1016/j.actbio.2013.01.026] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 01/09/2013] [Accepted: 01/24/2013] [Indexed: 02/02/2023]
Abstract
Human mesenchymal stem cell (hMSC) migration and recruitment play a critical role during bone fracture healing. Within the complex three-dimensional (3-D) in vivo microenvironment, hMSC migration is regulated through a myriad of extracellular cues. Here, we use a thiol-ene photopolymerized hydrogel to recapitulate structural and bioactive inputs in a tunable manner to understand their role in regulating 3-D hMSC migration. Specifically, peptide-functionalized poly(ethylene glycol) hydrogels were used to encapsulate hMSC while varying the crosslinking density, from 0.18±0.02 to 1.60±0.04 mM, and the adhesive ligand density, from 0.001 to 1.0 mM. Using live-cell videomicroscopy, migratory cell paths were tracked and fitted to a Persistent Random Walk model. It was shown that hMSC migrating through the lowest crosslinking density and highest adhesivity had more sustained polarization, higher migrating speeds (17.6±0.9 μm h(-1)) and higher cell spreading (elliptical form factor=3.9±0.2). However, manipulation of these material properties did not significantly affect migration persistence. Further, there was a monotonic increase in cell speed and spreading with increasing adhesivity that showed a lack of the biphasic trend seen in 2-D cell migration. Immunohistochemistry showed well-formed actin fibers and β1 integrin staining at the ends of stress fibers. This thiol-ene platform provides a highly tunable substrate to characterize 3-D hMSC migration that can be applied as an implantable cell carrier platform or for the recruitment of endogenous hMSC in vivo.
Collapse
|
226
|
Alamein MA, Liu Q, Stephens S, Skabo S, Warnke F, Bourke R, Heiner P, Warnke PH. Nanospiderwebs: artificial 3D extracellular matrix from nanofibers by novel clinical grade electrospinning for stem cell delivery. Adv Healthc Mater 2013. [PMID: 23184860 DOI: 10.1002/adhm.201200287] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Novel clinical grade electrospinning methods could provide three-dimensional (3D) nanostructured biomaterials comprising of synthetic or natural biopolymer nanofibers. Such advanced materials could potentially mimic the natural extracellular matrix (ECM) accurately and may provide superior niche-like spaces on the subcellular scale for optimal stem-cell attachment and individual cell homing in regenerative therapies. The goal of this study was to design several novel "nanofibrous extracellular matrices" (NF-ECMs) with a natural mesh-like 3D architecture through a unique needle-free multi-jet electrospinning method in highly controlled manner to comply with good manufacturing practices (GMP) for the production of advanced healthcare materials for regenerative medicine, and to test cellular behavior of human mesenchymal stem cells (HMSCs) on these. Biopolymers manufactured as 3D NF-ECM meshes under clinical grade GMP-like conditions show higher intrinsic cytobiocompatibility with superior cell integration and proliferation if compared to their 2D counterparts or a clinically-approved collagen membrane.
Collapse
Affiliation(s)
- Mohammad A Alamein
- Clem Jones Research Centre For Stem Cells & Tissue Regenerative Therapies, Bond University, Gold Coast, QLD, 4229, Australia
| | | | | | | | | | | | | | | |
Collapse
|
227
|
Cell transplantation as an initiator of endogenous stem cell-based tissue repair. Curr Opin Organ Transplant 2013; 17:670-4. [PMID: 23111645 DOI: 10.1097/mot.0b013e328359a617] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW The aim of this review is to summarize recent findings of endogenous cell-based tissue repair triggered by a multitude of approaches, especially stem cell therapy. RECENT FINDINGS The emerging evidence has demonstrated that an important role of transplanted cells is to act as an initiator to trigger endogenous stem cell-mediated tissue repair after injury. Multiple approaches may orchestrate endogenous regeneration. The low efficacy of endogenous stem cells may be due to deficiency of cytokines to activate and induce endogenous stem cell homing, relatively insufficient endogenous stem cell pool, diseases or aging-related dysfunction of endogenous stem cells, and hostile microenvironments that limit the capacity of endogenous stem cells to repair damaged tissue. In certain situations, external stimulation and/or exogenous stem cells may be required to catalyze the repair. SUMMARY Endogenous stem cells are playing an important role in tissue repair. An important role of transplanted cells is to act as an initiator to trigger endogenous stem cell-based tissue repair.
Collapse
|
228
|
Najafi R, Sharifi AM. Deferoxamine preconditioning potentiates mesenchymal stem cell homing in vitro and in streptozotocin-diabetic rats. Expert Opin Biol Ther 2013; 13:959-72. [PMID: 23536977 DOI: 10.1517/14712598.2013.782390] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Today, cell therapy is considered a promising alternative in treatment of several diseases such as type 1 diabetes. Loss of transplanted stem cell and more importantly scarcity in the number of cells reaching to target tissue is a major obstacle in cell therapy. There is evidences showing that deferoxamine (DFO), an iron chelator, increases the mobilization and homing of progenitor cells through increasing the stability of hypoxia-inducible factor 1α (HIF-1α) protein. In this study, the effect of DFO on some factors involved in homing of bone marrow-derived mesenchymal stem cell was investigated, and the other objectives of this research were to determine whether DFO is able to increase migration and subsequent homing of mesenchymal stem cell (MSCs) both in vitro and in vivo in streptozotocin-diabetic rats. RESEARCH DESIGN AND METHODS MSCs were treated by DFO in minimal essential medium α (αMEM) for 24 h. The expression and localization of HIF-1α were evaluated by western blotting and immunocytochemistry. The expression of C-X-C chemokine receptor type 4 (CXCR-4) and chemokine receptor 2 (CCR2) were assessed by western blotting and RT-PCR. The activity of matrix metalloproteinases (MMP) -2 and -9 were measured by gelatin zymography. Finally, in vitro migration of MSCs toward different concentrations of stromal cell-derived factor and monocyte chemotactic protein-1 were also evaluated. To demonstrate the homing of MSCs in vivo, DFO-treated chloromethyl-benzamidodialkylcarbocyanine-labeled MSCs were injected into the tail vein of rats, and the number of stained MSCs reaching to the pancreas were determined after 24 h. RESULTS In DFO-treated MSCs, expression of HIF-1α (p < 0.001), CXCR4 (p < 0.001), CCR2 (p < 0.001), and the activity of MMP-2 (p < 0.01) and MMP-9 (p < 0.05) were significantly increased compared to control groups. Elevation of HIF-1α, upregulation of CXCR4/CCR2 and higher activity of MMP-2/MMP-9 in DFO-treated MSCs were reversed by 2-methoxyestradiol (2-ME; 5 μmol), a HIF-1α inhibitor. The in vitro migrations as well as in vivo homing of DFO-treated MSCs were also significantly higher than control groups (p < 0.05). CONCLUSIONS Preconditioning of MSCs by DFO prior to transplantation could increase homing of MSCs through affecting some chemokine receptors as well as proteases involved and eventually improving the efficacy of cell therapy.
Collapse
Affiliation(s)
- R Najafi
- Tehran University of Medical Sciences, School of Medicine, Razi Drug Research Center, Department of Pharmacology, Tehran, Iran
| | | |
Collapse
|
229
|
Du Y, Yun H, Yang E, Schuman JS. Stem cells from trabecular meshwork home to TM tissue in vivo. Invest Ophthalmol Vis Sci 2013; 54:1450-9. [PMID: 23341019 DOI: 10.1167/iovs.12-11056] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE To investigate the potential of human trabecular meshwork stem cells (TMSCs) for homing to mouse TM tissue and survival in vivo. METHODS Human TMSCs and fibroblasts were labeled with fluorescent membrane dye DiO and injected into normal mouse anterior chamber. Stem cell and TM cell markers were identified by immunofluorescent staining of cryosections or tissue whole mounts. Apoptosis was determined by TUNEL assay. Replicating and inflammatory cells were detected by bromodeoxyuridine (BrdU) incorporation and anti-CD45 staining, respectively. Quantitative RT-PCR detected gene expression of injected cells after isolation by fluorescence activated cell sorting. Intraocular pressure was measured using a TonoLab rebound tonometer. RESULTS Expanded cultures of DiO-labeled TMSCs expressed stem cell markers preferentially in DiO positive cells, demonstrating a slow-cycling, label-retaining stem cell phenotype. DiO-labeled TMSCs injected into the anterior chamber of normal mice localized primarily in TM, remaining in the tissue at least 4 months. Within 1 week, TM-associated TMSCs began expressing TM marker protein CHI3L1. Fibroblasts injected in mouse anterior chamber showed distributed localization in corneal endothelium, lens epithelium, and TM and did not express CHI3L1. Little apoptosis was detected in injected TM tissue and intraocular pressure was not elevated during the experiment. Dividing cells or CD45-staining cells were not detected after TMSC-injection. CONCLUSIONS Stem cells isolated from human TM and expanded in vitro exhibit the ability to home to the TM and differentiate into TM cells in vivo. Such cells present a potential for development of a novel cell-based therapy for glaucoma.
Collapse
Affiliation(s)
- Yiqin Du
- UPMC Eye Center, Ophthalmology and Visual Science Research Center, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | | | | | | |
Collapse
|
230
|
Taschieri S, Corbella S, Tsesis I, Del Fabbro M. Impact of the use of plasma rich in growth factors (PRGF) on the quality of life of patients treated with endodontic surgery when a perforation of sinus membrane occurred. A comparative study. Oral Maxillofac Surg 2013; 18:43-52. [PMID: 23306948 DOI: 10.1007/s10006-012-0386-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 12/20/2012] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The aim of this retrospective investigation was to evaluate the postoperative quality of life after endodontic surgery in maxillary molars when a sinus membrane perforation occurred and platelet concentrates were used. MATERIALS AND METHODS Included patients were treated by microsurgical endodontic treatment in molar and premolar maxillary regions between 2007 and 2010. Patients who fulfilled the inclusion criteria were screened. Data from the quality of life questionnaire were analyzed. The use of plasma rich in growth factors (PRGF) (test group) was compared with a control group when a Schneiderian membrane perforation occurred during endodontic surgery performed with a modern technique in maxillary molars and premolars. RESULTS A total of 20 patients (12 in the control group and eight in the test group) fulfilled the inclusion criteria. No differences were evaluated at baseline for clinical parameters. Significantly improved patients' quality of life was observed in the test group considering symptoms as swelling, bad breath or taste, and pain. Functional activities were less impaired in the test group and swelling was significantly higher in the control group. In the test group, pain was significantly lower than the control group during the first 6 days after surgery and also, the consumption of painkillers was lower for patients belonging to the test group even if it was not statistically significant. DISCUSSION In general, a small sinus membrane perforation (less than 6 mm) during endodontic surgery did not cause severe complications. The use of platelet concentrates could be effective in reducing the impact on patients' quality of life, decreasing pain and surgery side effects as well as swelling.
Collapse
Affiliation(s)
- S Taschieri
- Department of Biomedical, Surgical and Dental Sciences, IRCCS Istituto Ortopedico Galeazzi, Università degli Studi di Milano, Milan, Italy
| | | | | | | |
Collapse
|
231
|
Ahmed LA. Stem cells and cardiac repair: alternative and multifactorial approaches. ACTA ACUST UNITED AC 2013. [DOI: 10.7243/2050-1218-2-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
232
|
Saravanakumar M, Devaraj H. Distribution and homing pattern of c-kit+ Sca-1+ CXCR4+ resident cardiac stem cells in neonatal, postnatal, and adult mouse heart. Cardiovasc Pathol 2012; 22:257-63. [PMID: 23273785 DOI: 10.1016/j.carpath.2012.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Revised: 11/09/2012] [Accepted: 11/13/2012] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION The origin of heart-forming cells and their roles in organ development have fascinated biologists for over a century. C-X-C chemokine receptor type 4 plays a crucial role during embryonic development and in maintaining the stem cell niche and homing. The aim of the present was to study the expression pattern of resident cardiac stem cell markers and their homing factor in neonatal, postnatal, and adult mouse heart. METHODS Cardiac stem cell protein expression was analyzed using immunofluorescence, immunohistochemistry, and Western blotting. The messenger ribonucleic acid expression of cardiac stem cell markers c-kit, stem cell antigen-1, and homing factor C-X-C chemokine receptor type 4 was quantitatively analyzed using quantitative polymerase chain reaction. Data were analyzed using Student's t test and two-way analysis using SPSS software. RESULTS Stem cell antigen-1- and c-kit-positive cell populations were heterogeneously distributed in the adult and postnatal hearts but scattered in the neonatal heart. The expression of c-kit showed a significant difference between right and left atrium, though it was higher compared to ventricles. The homing factor C-X-C chemokine receptor type 4 expression was higher in the neonatal heart than in the postnatal heart but was not detectable in the adult heart. CONCLUSIONS The present study reveals the distribution of cardiac stem cells in the different compartments of the heart and significant reduction in their number in adult heart. Cardiac stem cells are higher in the atrium than in the ventricle, suggesting the atria as the source of cardiac stem cell.
Collapse
Affiliation(s)
- Marimuthu Saravanakumar
- Unit of Biochemistry, Department of Zoology, University of Madras, School of Life Science, Maraimalai Campus, Guindy, Chennai-600 025, Tamil Nadu, India
| | | |
Collapse
|
233
|
Minguell JJ, Allers C, Lasala GP. Mesenchymal stem cells and the treatment of conditions and diseases: the less glittering side of a conspicuous stem cell for basic research. Stem Cells Dev 2012; 22:193-203. [PMID: 23025629 DOI: 10.1089/scd.2012.0417] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Not too long ago, several motivated and forward-looking articles were published describing the cellular and molecular properties of mesenchymal stem cells (MSCs), specially highlighting their potential for self-renewal, commitment, differentiation, and maturation into specific mesoderm-derived lineages. A very influential publication of that period entitled "Mesenchymal stem cells: No longer second class marrow citizens" [1] raised the point of view that "…challenges to harness MSC cell therapy to treat diseases … need to wait for the full comprehension that marrow is a rich source of mesenchyme-derived cells whose potential is still far from fully appreciated." Whether or not the prophecy of Gerson was fulfilled, in the last 8 years it has become evident that infusing MSCs into patients suffering a variety of disorders represents a viable option for medical treatment. Accordingly, a vast number of articles have explored the privileged cellular and molecular features of MSCs prepared from sources other than the canonical, represented by the bone marrow. This review will provide more information neither related to the biological attractiveness of MSCs nor to the success after their clinical use. Rather, we would like to underscore several "critical and tangential" issues, not always discussed in biomedical publications, but relevant to the clinical utilization of bone-marrow-derived MSCs.
Collapse
Affiliation(s)
- Jose J Minguell
- TCA Cellular Therapy, 101 Judge Tanner Boulevard, Covington, LA 70433, USA.
| | | | | |
Collapse
|
234
|
Ji W, Yang F, Ma J, Bouma MJ, Boerman OC, Chen Z, van den Beucken JJJP, Jansen JA. Incorporation of stromal cell-derived factor-1α in PCL/gelatin electrospun membranes for guided bone regeneration. Biomaterials 2012; 34:735-45. [PMID: 23117215 DOI: 10.1016/j.biomaterials.2012.10.016] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 10/08/2012] [Indexed: 01/07/2023]
Abstract
The goal of this work was to evaluate the effect of membrane functionalization with a chemotactic factor on cell recruitment and bone formation in order to develop a bioactive membrane for guided bone regeneration (GBR) applications. To this end, GBR membranes were prepared by electrospinning using poly(ε-caprolactone) (PCL) blended with type B-gelatin, and functionalized with stromal cell derived factor-1α (SDF-1α) via physical adsorption. Firstly, the obtained membranes were evaluated in vitro for SDF-1α release and chemotactic effect on bone marrow stromal cells (BMSCs). Subsequently, in vivo BMSCs recruitment and bone regeneration in response to SDF-1α loaded PCL/gelatin electrospun membranes were assessed in rat cranial defects. The results showed that PCL/gelatin electrospun membranes provided a diffusion-controlled SDF-1α release profile. Furthermore, the membranes loaded with different amounts of SDF-1α (50-400 ng) significantly induced stimulated chemotactic migration of BMSCs in vitro without dose-dependent effects. Eight weeks after implantation in rat cranial defects, SDF-1α loaded membranes yielded a 6-fold increase in the amount of bone formation compared to the bare membranes, albeit that contribution of in vivo BMSCs recruitment to the bone regeneration could not be ascertained. In conclusion, the results of current study indicate the potential for using SDF-1α loaded PCL/gelatin electrospun membrane as a bioactive membrane, which is beneficial for optimizing clinical application of GBR strategies.
Collapse
Affiliation(s)
- Wei Ji
- Department of Biomaterials, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
235
|
Lu H, Xie C, Zhao YM, Chen FM. Translational research and therapeutic applications of stem cell transplantation in periodontal regenerative medicine. Cell Transplant 2012; 22:205-29. [PMID: 23031442 DOI: 10.3727/096368912x656171] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Stem cells have received a great deal of interest from the research community as potential therapeutic "tools" for a variety of chronic debilitating diseases that lack clinically effective therapies. Stem cells are also of interest for the regeneration of tooth-supporting tissues that have been lost to periodontal disease. Indeed, substantial data have demonstrated that the exogenous administration of stem cells or their derivatives in preclinical animal models of periodontal defects can restore damaged tissues to their original form and function. As we discuss here, however, considerable hurdles must be overcome before these findings can be responsibly translated to novel clinical therapies. Generally, the application of stem cells for periodontal therapy in clinics will not be realized until the best cell(s) to use, the optimal dose, and an effective mode of administration are identified. In particular, we need to better understand the mechanisms of action of stem cells after transplantation in the periodontium and to learn how to preciously control stem cell fates in the pathological environment around a tooth. From a translational perspective, we outline the challenges that may vary across preclinical models for the evaluation of stem cell therapy in situations that require periodontal reconstruction and the safety issues that are related to clinical applications of human stem cells. Although clinical trials that use autologous periodontal ligament stem cells have been approved and have already been initiated, proper consideration of the technical, safety, and regulatory concerns may facilitate, rather than inhibit, the clinical translation of new therapies.
Collapse
Affiliation(s)
- Hong Lu
- Department of Periodontology and Oral Medicine, School of Stomatology, Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | | | | | | |
Collapse
|
236
|
The effect of aging on the pluripotential capacity and regenerative potential of human periodontal ligament stem cells. Biomaterials 2012; 33:6974-86. [DOI: 10.1016/j.biomaterials.2012.06.032] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2012] [Accepted: 06/17/2012] [Indexed: 12/14/2022]
|
237
|
McNulty MA, Virdi AS, Christopherson KW, Sena K, Frank RR, Sumner DR. Adult stem cell mobilization enhances intramembranous bone regeneration: a pilot study. Clin Orthop Relat Res 2012; 470:2503-12. [PMID: 22528386 PMCID: PMC3830081 DOI: 10.1007/s11999-012-2357-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Stem cell mobilization, which is defined as the forced egress of stem cells from the bone marrow to the peripheral blood (PB) using chemokine receptor agonists, is an emerging concept for enhancing tissue regeneration. However, the effect of stem cell mobilization by a single injection of the C-X-C chemokine receptor type 4 (CXCR4) antagonist AMD3100 on intramembranous bone regeneration is unclear. QUESTIONS/PURPOSES We therefore asked: Does AMD3100 mobilize adult stem cells in C57BL/6 mice? Are stem cells mobilized to the PB after marrow ablation? And does AMD3100 enhance bone regeneration? METHODS Female C57BL/6 mice underwent femoral marrow ablation surgery alone (n = 25), systemic injection of AMD3100 alone (n = 15), or surgery plus AMD3100 (n = 57). We used colony-forming unit assays, flow cytometry, and micro-CT to investigate mobilization of mesenchymal stem cells, endothelial progenitor cells, and hematopoietic stem cells to the PB and bone regeneration. RESULTS AMD3100 induced mobilization of stem cells to the PB, resulting in a 40-fold increase in mesenchymal stem cells. The marrow ablation injury mobilized all three cell types to the PB over time. Administration of AMD3100 led to a 60% increase in bone regeneration at Day 21. CONCLUSIONS A single injection of a CXCR4 antagonist lead to stem cell mobilization and enhanced bone volume in the mouse marrow ablation model of intramembranous bone regeneration.
Collapse
Affiliation(s)
- Margaret A. McNulty
- Department of Anatomy & Cell Biology, Rush University Medical Center, 600 Paulina Street, Chicago, IL 60612 USA
| | - Amarjit S. Virdi
- Department of Anatomy & Cell Biology, Rush University Medical Center, 600 Paulina Street, Chicago, IL 60612 USA
| | | | - Kotaro Sena
- Department of Anatomy & Cell Biology, Rush University Medical Center, 600 Paulina Street, Chicago, IL 60612 USA
| | - Robin R. Frank
- Division of Hematology & Oncology, Rush University Medical Center, Chicago, IL USA
| | - Dale R. Sumner
- Department of Anatomy & Cell Biology, Rush University Medical Center, 600 Paulina Street, Chicago, IL 60612 USA
| |
Collapse
|
238
|
Deveza L, Choi J, Yang F. Therapeutic angiogenesis for treating cardiovascular diseases. Theranostics 2012; 2:801-14. [PMID: 22916079 PMCID: PMC3425124 DOI: 10.7150/thno.4419] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 05/29/2012] [Indexed: 12/30/2022] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide and is often associated with partial or full occlusion of the blood vessel network in the affected organs. Restoring blood supply is critical for the successful treatment of cardiovascular diseases. Therapeutic angiogenesis provides a valuable tool for treating cardiovascular diseases by stimulating the growth of new blood vessels from pre-existing vessels. In this review, we discuss strategies developed for therapeutic angiogenesis using single or combinations of biological signals, cells and polymeric biomaterials. Compared to direct delivery of growth factors or cells alone, polymeric biomaterials provide a three-dimensional drug-releasing depot that is capable of facilitating temporally and spatially controlled release. Biomimetic signals can also be incorporated into polymeric scaffolds to allow environmentally-responsive or cell-triggered release of biological signals for targeted angiogenesis. Recent progress in exploiting genetically engineered stem cells and endogenous cell homing mechanisms for therapeutic angiogenesis is also discussed.
Collapse
|
239
|
Mirabella T, Hartinger J, Lorandi C, Gentili C, van Griensven M, Cancedda R. Proangiogenic Soluble Factors from Amniotic Fluid Stem Cells Mediate the Recruitment of Endothelial Progenitors in a Model of Ischemic Fasciocutaneous Flap. Stem Cells Dev 2012; 21:2179-88. [DOI: 10.1089/scd.2011.0639] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
- Teodelinda Mirabella
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
- AOU San Martino—IST National Institute for Cancer Research, Genova, Italy
| | - Joachim Hartinger
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Centre, Vienna, Austria
| | - Christian Lorandi
- Department of Materials Engineering and Industrial Technologies, University of Trento, Trento, Italy
| | - Chiara Gentili
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
- AOU San Martino—IST National Institute for Cancer Research, Genova, Italy
| | - Martijn van Griensven
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Centre, Vienna, Austria
| | - Ranieri Cancedda
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
- AOU San Martino—IST National Institute for Cancer Research, Genova, Italy
| |
Collapse
|
240
|
Niu LN, Jiao K, Qi YP, Nikonov S, Yiu CKY, Arola DD, Gong SQ, El-Marakby A, Carrilho MRO, Hamrick MW, Hargreaves KM, Diogenes A, Chen JH, Pashley DH, Tay FR. Intrafibrillar silicification of collagen scaffolds for sustained release of stem cell homing chemokine in hard tissue regeneration. FASEB J 2012; 26:4517-29. [PMID: 22859369 DOI: 10.1096/fj.12-210211] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Traditional bone regeneration strategies relied on supplementation of biomaterials constructs with stem or progenitor cells or growth factors. By contrast, cell homing strategies employ chemokines to mobilize stem or progenitor cells from host bone marrow and tissue niches to injured sites. Although silica-based biomaterials exhibit osteogenic and angiogenic potentials, they lack cell homing capability. Stromal cell-derived factor-1 (SDF-1) plays a pivotal role in mobilization and homing of stem cells to injured tissues. In this work, we demonstrated that 3-dimensional collagen scaffolds infiltrated with intrafibrillar silica are biodegradable and highly biocompatible. They exhibit improved compressive stress-strain responses and toughness over nonsilicified collagen scaffolds. They are osteoconductive and up-regulate expressions of osteogenesis- and angiogenesis-related genes more significantly than nonsilicified collagen scaffolds. In addition, these scaffolds reversibly bind SDF-1α for sustained release of this chemokine, which exhibits in vitro cell homing characteristics. When implanted subcutaneously in an in vivo mouse model, SDF-1α-loaded silicified collagen scaffolds stimulate the formation of ectopic bone and blood capillaries within the scaffold and abrogate the need for cell seeding or supplementation of osteogenic and angiogenic growth factors. Intrafibrillar-silicified collagen scaffolds with sustained SDF-1α release represent a less costly and complex alternative to contemporary cell seeding approaches and provide new therapeutic options for in situ hard tissue regeneration.
Collapse
Affiliation(s)
- Li-Na Niu
- School of Stomatology, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
241
|
Liu B, Tan XY, Liu YP, Xu XF, Li L, Xu HY, An R, Chen FM. The adjuvant use of stromal vascular fraction and platelet-rich fibrin for autologous adipose tissue transplantation. Tissue Eng Part C Methods 2012; 19:1-14. [PMID: 22681647 DOI: 10.1089/ten.tec.2012.0126] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Autologous adipose transplantation is rapidly gaining popularity for the restoration of soft tissue defects and lipoatrophy as well as for aesthetic improvements (e.g., facial reconstruction and rejuvenation). However, the current technique is crude that suffers from serious demerits, particularly the long-term unpredictability of volume maintenance due to resorption of the grafted adipose tissue and limited adipogenesis. We hypothesized that the adjuvant use of patient-derived adipose stromal vascular fraction (SVF) and platelet-rich fibrin (PRF) may enhance the overall outcome of autologous fat grafting in plastic and reconstructive surgery. Autologous SVF, with a mean cell number of (4.8±3.79)×10⁷ cells/mL and a mean cell viability of 71.8%, and autologous PRF, with sustained release of multiple angiogenic growth factors, were created before surgical use. The following adipose tissue implants were injected subcutaneously into a rabbit ear's auricula according to the following study design: 2 mL adipose granules and 0.2 mL normal saline solution (AG+NS group), 2 mL adipose granules and 0.2 mL SVF (AG+SVF group), 2 mL adipose granules and 0.2 mL PRF (AG+PRF group), or 2 mL adipose granules combined with 0.1 mL SVF and 0.1 mL PRF (AG+SVF+PRF group). Histological examinations showed that the implanted adipose granules were well engrafted in the AG+SVF+PRF group, with a higher microvessel density 4 weeks postimplantation compared with the other three groups (p<0.01). Twenty-four weeks postimplantation, the resorption rates of implanted tissue in each group were 49.39%±9.47%, 27.25%±4.37%, 36.41%±8.47%, and 17.37%±6.22%, respectively, and were significantly different (p<0.01). The results demonstrated that the efficacy of adipose tissue implantation can be enhanced by using autologous PRF and SVF as therapeutic adjuvants, offering a clinically translatable strategy for soft tissue augmentation and reconstruction.
Collapse
Affiliation(s)
- Bin Liu
- Department of Oral Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | | | | | | | | | | | | | | |
Collapse
|
242
|
Zhang Y, Cheng N, Miron R, Shi B, Cheng X. Delivery of PDGF-B and BMP-7 by mesoporous bioglass/silk fibrin scaffolds for the repair of osteoporotic defects. Biomaterials 2012; 33:6698-708. [PMID: 22763224 DOI: 10.1016/j.biomaterials.2012.06.021] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Accepted: 06/15/2012] [Indexed: 01/23/2023]
Abstract
Osteoporosis is a chronic disease affecting millions of people worldwide caused by an imbalance between bone-forming osteoblasts and bone-resorbing osteoclasts. Despite recent developments in pharmacological agents to prevent osteoporotic-related fractures, much less attention has been placed on the repair of bone defects following fracture. Critical to this process is the recruitment of mesenchymal stem cells (MSCs) to defect sites by growth factors. One method which has been effective for the sustained release of growth factors is that of gene therapy. The aim of the present study was to investigate newly developed mesoporous bioglass/silk fibrin scaffolds containing adPDGF-b and adBMP-7 into osteoporotic critical-sized femur defects in ovariectomised rats following treatment periods of 2 and 4 weeks. In vivo osteogenetic efficiency evaluated by μ-CT analysis, hematoxylin and eosin staining, and immunohistochemical (type I collagen, osteopontin and BSP) revealed significantly new bone formation in defects containing adenovirus for both PDGF-b and BMP-7 when compared to scaffolds alone and scaffolds containing BMP-7. TRAP-positive staining also demonstrated the ability for these scaffolds to be degraded over time and initiate bone turnover/remodeling. Although the use of gene therapy for clinical applications is still in its infancy, results from the present study demonstrate their potent ability to recruit mesenchymal progenitor cells through sustained release of PDGF-b and BMP-7 which may be beneficial for patients suffering from osteoporotic-related fractures.
Collapse
Affiliation(s)
- Yufeng Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan 430079, PR China.
| | | | | | | | | |
Collapse
|
243
|
Bottino MC, Thomas V, Schmidt G, Vohra YK, Chu TMG, Kowolik MJ, Janowski GM. Recent advances in the development of GTR/GBR membranes for periodontal regeneration—A materials perspective. Dent Mater 2012; 28:703-21. [DOI: 10.1016/j.dental.2012.04.022] [Citation(s) in RCA: 368] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 02/21/2012] [Accepted: 04/16/2012] [Indexed: 10/28/2022]
|
244
|
Chen FM, Sun HH, Lu H, Yu Q. Stem cell-delivery therapeutics for periodontal tissue regeneration. Biomaterials 2012; 33:6320-44. [PMID: 22695066 DOI: 10.1016/j.biomaterials.2012.05.048] [Citation(s) in RCA: 211] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 05/20/2012] [Indexed: 02/07/2023]
Abstract
Periodontitis, an inflammatory disease, is the most common cause of tooth loss in adults. Attempts to regenerate the complex system of tooth-supporting apparatus (i.e., the periodontal ligament, alveolar bone and root cementum) after loss/damage due to periodontitis have made some progress recently and provide a useful experimental model for the evaluation of future regenerative therapies. Concentrated efforts have now moved from the use of guided tissue/bone regeneration technology, a variety of growth factors and various bone grafts/substitutes toward the design and practice of endogenous regenerative technology by recruitment of host cells (cell homing) or stem cell-based therapeutics by transplantation of outside cells to enhance periodontal tissue regeneration and its biomechanical integration. This shift is driven by the general inability of conventional therapies to deliver satisfactory outcomes, particularly in cases where the disease has caused large tissue defects in the periodontium. Cell homing and cell transplantation are both scientifically meritorious approaches that show promise to completely and reliably reconstitute all tissue and connections damaged through periodontal disease, and hence research into both directions should continue. In view of periodontal regeneration by paradigms that unlock the body's innate regenerative potential has been reviewed elsewhere, this paper specifically explores and analyses the stem cell types and cell delivery strategies that have been or have the potential to be used as therapeutics in periodontal regenerative medicine, with particular emphasis placed on the efficacy and safety concerns of current stem cell-based periodontal therapies that may eventually enter into the clinic.
Collapse
Affiliation(s)
- Fa-Ming Chen
- Department of Periodontology and Oral Medicine, School of Stomatology, Fourth Military Medical University, Xi'an 710032, Shaanxi, PR China.
| | | | | | | |
Collapse
|
245
|
Polycaprolactone electrospun mesh conjugated with an MSC affinity peptide for MSC homing in vivo. Biomaterials 2012; 33:3375-87. [DOI: 10.1016/j.biomaterials.2012.01.033] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Accepted: 01/14/2012] [Indexed: 12/13/2022]
|
246
|
Jakob F, Ebert R, Rudert M, Nöth U, Walles H, Docheva D, Schieker M, Meinel L, Groll J. In situ guided tissue regeneration in musculoskeletal diseases and aging : Implementing pathology into tailored tissue engineering strategies. Cell Tissue Res 2012; 347:725-35. [PMID: 22011785 PMCID: PMC3306563 DOI: 10.1007/s00441-011-1237-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 09/01/2011] [Indexed: 12/17/2022]
Abstract
In situ guided tissue regeneration, also addressed as in situ tissue engineering or endogenous regeneration, has a great potential for population-wide "minimal invasive" applications. During the last two decades, tissue engineering has been developed with remarkable in vitro and preclinical success but still the number of applications in clinical routine is extremely small. Moreover, the vision of population-wide applications of ex vivo tissue engineered constructs based on cells, growth and differentiation factors and scaffolds, must probably be deemed unrealistic for economic and regulation-related issues. Hence, the progress made in this respect will be mostly applicable to a fraction of post-traumatic or post-surgery situations such as big tissue defects due to tumor manifestation. Minimally invasive procedures would probably qualify for a broader application and ideally would only require off the shelf standardized products without cells. Such products should mimic the microenvironment of regenerating tissues and make use of the endogenous tissue regeneration capacities. Functionally, the chemotaxis of regenerative cells, their amplification as a transient amplifying pool and their concerted differentiation and remodeling should be addressed. This is especially important because the main target populations for such applications are the elderly and diseased. The quality of regenerative cells is impaired in such organisms and high levels of inhibitors also interfere with regeneration and healing. In metabolic bone diseases like osteoporosis, it is already known that antagonists for inhibitors such as activin and sclerostin enhance bone formation. Implementing such strategies into applications for in situ guided tissue regeneration should greatly enhance the efficacy of tailored procedures in the future.
Collapse
Affiliation(s)
- Franz Jakob
- Orthopedic Center for Musculoskeletal Research, Julius Maximilians University of Wuerzburg, Brettreichstrasse 11, D-97082 Wuerzburg, Germany
| | - Regina Ebert
- Orthopedic Center for Musculoskeletal Research, Julius Maximilians University of Wuerzburg, Brettreichstrasse 11, D-97082 Wuerzburg, Germany
| | - Maximilian Rudert
- Orthopedic Center for Musculoskeletal Research, Julius Maximilians University of Wuerzburg, Brettreichstrasse 11, D-97082 Wuerzburg, Germany
| | - Ulrich Nöth
- Orthopedic Center for Musculoskeletal Research, Julius Maximilians University of Wuerzburg, Brettreichstrasse 11, D-97082 Wuerzburg, Germany
| | - Heike Walles
- Institute for Tissue Engineering and Regenerative Medicine, Julius Maximilians University of Wuerzburg, Röntgenring 11, D-97070 Wuerzburg, Germany
| | - Denitsa Docheva
- Experimental Surgery and Regenerative Medicine, Ludwig Maximilians University Munich, Nußbaumstrasse 20, D-80336 München, Germany
| | - Matthias Schieker
- Experimental Surgery and Regenerative Medicine, Ludwig Maximilians University Munich, Nußbaumstrasse 20, D-80336 München, Germany
| | - Lorenz Meinel
- Chair for Pharmaceutical Technology, Julius Maximilians University of Wuerzburg, Am Hubland, D-97074 Wuerzburg, Germany
| | - Jürgen Groll
- Department and Chair of Functional Materials in Medicine and Dentistry, Julius Maximilians University of Wuerzburg, Pleicherwall 2, D-97070 Wuerzburg, Germany
| |
Collapse
|
247
|
Lim SY, Hausenloy DJ. Remote ischemic conditioning: from bench to bedside. Front Physiol 2012; 3:27. [PMID: 22363297 PMCID: PMC3282534 DOI: 10.3389/fphys.2012.00027] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 02/04/2012] [Indexed: 12/13/2022] Open
Abstract
Remote ischemic conditioning (RIC) is a therapeutic strategy for protecting organs or tissue against the detrimental effects of acute ischemia-reperfusion injury (IRI). It describes an endogenous phenomenon in which the application of one or more brief cycles of non-lethal ischemia and reperfusion to an organ or tissue protects a remote organ or tissue from a sustained episode of lethal IRI. Although RIC protection was first demonstrated to protect the heart against acute myocardial infarction, its beneficial effects are also seen in other organs (lung, liver, kidney, intestine, brain) and tissues (skeletal muscle) subjected to acute IRI. The recent discovery that RIC can be induced non-invasively by simply inflating and deflating a standard blood pressure cuff placed on the upper arm or leg, has facilitated its translation into the clinical setting, where it has been reported to be beneficial in a variety of cardiac scenarios. In this review article we provide an overview of RIC, the potential underlying mechanisms, and its potential as a novel therapeutic strategy for protecting the heart and other organs from acute IRI.
Collapse
Affiliation(s)
- Shiang Yong Lim
- O'Brien Institute, Department of Surgery, University of Melbourne, St Vincent's Hospital Melbourne, VIC, Australia
| | | |
Collapse
|
248
|
Exploiting extracellular matrix-stem cell interactions: A review of natural materials for therapeutic muscle regeneration. Biomaterials 2012; 33:428-43. [DOI: 10.1016/j.biomaterials.2011.09.078] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 09/28/2011] [Indexed: 02/07/2023]
|
249
|
Colonization of recipient tissues with transplanted murine bone marrow cells. Transfus Apher Sci 2011; 46:109-11. [PMID: 22169382 DOI: 10.1016/j.transci.2011.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 11/17/2011] [Indexed: 11/21/2022]
|
250
|
Chen FM, Zhao YM, Jin Y, Shi S. Prospects for translational regenerative medicine. Biotechnol Adv 2011; 30:658-72. [PMID: 22138411 DOI: 10.1016/j.biotechadv.2011.11.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 11/12/2011] [Accepted: 11/15/2011] [Indexed: 02/06/2023]
Abstract
Translational medicine is an evolutional concept that encompasses the rapid translation of basic research for use in clinical disease diagnosis, prevention and treatment. It follows the idea "from bench to bedside and back", and hence relies on cooperation between laboratory research and clinical care. In the past decade, translational medicine has received unprecedented attention from scientists and clinicians and its fundamental principles have penetrated throughout biomedicine, offering a sign post that guides modern medical research toward a patient-centered focus. Translational regenerative medicine is still in its infancy, and significant basic research investment has not yet achieved satisfactory clinical outcomes for patients. In particular, there are many challenges associated with the use of cell- and tissue-based products for clinical therapies. This review summarizes the transformation and global progress in translational medicine over the past decade. The current obstacles and opportunities in translational regenerative medicine are outlined in the context of stem cell therapy and tissue engineering for the safe and effective regeneration of functional tissue. This review highlights the requirement for multi-disciplinary and inter-disciplinary cooperation to ensure the development of the best possible regenerative therapies within the shortest timeframe possible for the greatest patient benefit.
Collapse
Affiliation(s)
- Fa-Ming Chen
- Department of Periodontology & Oral Medicine, School of Stomatology, Fourth Military Medical University, Xi'an 710032, Shaanxi, PR China.
| | | | | | | |
Collapse
|