Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Nat Rev Rheumatol ; 19(7): 403-416, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37296196

ABSTRACT

Osteoarthritis (OA) is a disabling condition that affects billions of people worldwide and places a considerable burden on patients and on society owing to its prevalence and economic cost. As cartilage injuries are generally associated with the progressive onset of OA, robustly effective approaches for cartilage regeneration are necessary. Despite extensive research, technical development and clinical experimentation, no current surgery-based, material-based, cell-based or drug-based treatment can reliably restore the structure and function of hyaline cartilage. This paucity of effective treatment is partly caused by a lack of fundamental understanding of why articular cartilage fails to spontaneously regenerate. Thus, research studies that investigate the mechanisms behind the cartilage regeneration processes and the failure of these processes are critical to instruct decisions about patient treatment or to support the development of next-generation therapies for cartilage repair and OA prevention. This Review provides a synoptic and structured analysis of the current hypotheses about failure in cartilage regeneration, and the accompanying therapeutic strategies to overcome these hurdles, including some current or potential approaches to OA therapy.


Subject(s)
Cartilage, Articular , Osteoarthritis , Humans , Chondrocytes , Osteoarthritis/therapy , Regeneration
2.
Biomaterials ; 285: 121547, 2022 06.
Article in English | MEDLINE | ID: mdl-35533445

ABSTRACT

Targeting stem cells to cartilage lesions has the potential to enhance engraftment and chondrogenesis. Denatured type II collagen fibrils (gelatin) are exposed in lesions at the surface of osteoarthritic articular cartilage and are therefore ideal target sites. We have designed and investigated chimeric mutants of the three modules of the MMP-2 collagen binding domain (CBD) as potential ligands for stem cell targeting. We expressed full-length CBD for the first time and used it to identify the most important amino acid residues for binding to gelatin. Module 2 of CBD had the highest affinity binding to both Type I and Type II gelatin, whereas module 1 showed specificity for type II gelatin and module 3 for type I gelatin. We went on to generate chimeric forms of CBD consisting of three repeats of module 1 (111), module 2 (222) or module 3 (333). 111 lacked solubility and could not be further characterised. However 222 was found to bind to type II gelatin 14 times better than CBD, suggesting it would be optimal for attachment to cartilage lesions, whilst 333 was found to bind to type I gelatin 12 times better than CBD, suggesting it would be optimal for attachment to lesions in type I collagen-rich tissues. We coated 222 onto the external membrane of Mesenchymal Stem Cells and demonstrated higher attachment of the coated cells to type II gelatin than uncoated cells. We conclude that the three modules of CBD each have specific biological properties that can be exploited for targeting stem cells to cartilage lesions and other pathological sites.


Subject(s)
Cartilage, Articular , Matrix Metalloproteinase 2 , Carrier Proteins/metabolism , Cartilage/metabolism , Cartilage, Articular/metabolism , Collagen Type I/metabolism , Gelatin , Matrix Metalloproteinase 2/metabolism , Membranes, Artificial , Protein Binding , Protein Structure, Tertiary , Stem Cells/metabolism
3.
Stem Cells ; 38(11): 1438-1453, 2020 11.
Article in English | MEDLINE | ID: mdl-32652878

ABSTRACT

Mesenchymal stem cells (MSCs) have been investigated as a potential injectable therapy for the treatment of knee osteoarthritis, with some evidence of success in preliminary human trials. However, optimization and scale-up of this therapeutic approach depends on the identification of functional markers that are linked to their mechanism of action. One possible mechanism is through their chondrogenic differentiation and direct role in neo-cartilage synthesis. Alternatively, they could remain undifferentiated and act through the release of trophic factors that stimulate endogenous repair processes within the joint. Here, we show that extensive in vitro aging of bone marrow-derived human MSCs leads to loss of chondrogenesis but no reduction in trophic repair, thereby separating out the two modes of action. By integrating transcriptomic and proteomic data using Ingenuity Pathway Analysis, we found that reduced chondrogenesis with passage is linked to downregulation of the FOXM1 signaling pathway while maintenance of trophic repair is linked to CXCL12. In an attempt at developing functional markers of MSC potency, we identified loss of mRNA expression for MMP13 as correlating with loss of chondrogenic potential of MSCs and continued secretion of high levels of TIMP1 protein as correlating with the maintenance of trophic repair capacity. Since an allogeneic injectable osteoar therapy would require extensive cell expansion in vitro, we conclude that early passage MMP13+ , TIMP1-secretinghigh MSCs should be used for autologous OA therapies designed to act through engraftment and chondrogenesis, while later passage MMP13- , TIMP1-secretinghigh MSCs could be exploited for allogeneic OA therapies designed to act through trophic repair.


Subject(s)
Matrix Metalloproteinase 13/metabolism , Mesenchymal Stem Cell Transplantation/methods , Osteoarthritis/therapy , Tissue Engineering/methods , Tissue Inhibitor of Metalloproteinase-1/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism
4.
J Tissue Eng Regen Med ; 12(6): 1402-1411, 2018 06.
Article in English | MEDLINE | ID: mdl-29726103

ABSTRACT

Bioreactor systems will likely play a key role in establishing regulatory compliant and cost-effective production systems for manufacturing engineered tissue grafts for clinical applications. However, the automation of bioreactor systems could become considerably more complex and costly due to the requirements for additional storage and liquid handling technologies if unstable supplements are added to the culture medium. Ascorbic acid (AA) is a bioactive supplement that is commonly presumed to be essential for the generation of engineered cartilage tissues. However, AA can be rapidly oxidized and degraded. In this work, we addressed whether human nasal chondrocytes can redifferentiate, undergo chondrogenesis, and generate a cartilaginous extracellular matrix when cultured in the absence of AA. We found that when chondrocytes were cultured in 3D micromass pellets either with or without AA, there were no significant differences in their chondrogenic capacity in terms of gene expression or the amount of glycosaminoglycans. Moreover, 3D pellets cultured without AA contained abundant collagen Types II and I extracellular matrix. Although the amounts of Collagens II and I were significantly lower (34% and 50% lower) than in pellets cultured with AA, collagen fibers had similar thicknesses and distributions for both groups, as shown by scanning electron microscopy imaging. Despite the reduced amounts of collagen, if engineered cartilage grafts can be generated with sufficient properties that meet defined quality criteria without the use of unstable supplements such as AA, bioreactor automation requirements can be greatly simplified, thereby facilitating the development of more compact, user-friendly, and cost-effective bioreactor-based manufacturing systems.


Subject(s)
Ascorbic Acid/pharmacology , Cell Differentiation/drug effects , Chondrocytes/cytology , Chondrogenesis , Adult , Cell Survival/drug effects , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrogenesis/drug effects , Chondrogenesis/genetics , Collagen/metabolism , Culture Media , Extracellular Matrix/metabolism , Gene Expression Regulation/drug effects , Glycosaminoglycans/metabolism , Humans , Middle Aged , Young Adult
5.
Stem Cells ; 35(11): 2280-2291, 2017 11.
Article in English | MEDLINE | ID: mdl-28833807

ABSTRACT

Multipotent mesenchymal stem cells (MSCs) have enormous potential in tissue engineering and regenerative medicine. However, until now, their development for clinical use has been severely limited as they are a mixed population of cells with varying capacities for lineage differentiation and tissue formation. Here, we identify receptor tyrosine kinase-like orphan receptor 2 (ROR2) as a cell surface marker expressed by those MSCs with an enhanced capacity for cartilage formation. We generated clonal human MSC populations with varying capacities for chondrogenesis. ROR2 was identified through screening for upregulated genes in the most chondrogenic clones. When isolated from uncloned populations, ROR2+ve MSCs were significantly more chondrogenic than either ROR2-ve or unfractionated MSCs. In a sheep cartilage-repair model, they produced significantly more defect filling with no loss of cartilage quality compared with controls. ROR2+ve MSCs/perivascular cells were present in developing human cartilage, adult bone marrow, and adipose tissue. Their frequency in bone marrow was significantly lower in patients with osteoarthritis (OA) than in controls. However, after isolation of these cells and their initial expansion in vitro, there was greater ROR2 expression in the population derived from OA patients compared with controls. Furthermore, osteoarthritis-derived MSCs were better able to form cartilage than MSCs from control patients in a tissue engineering assay. We conclude that MSCs expressing high levels of ROR2 provide a defined population capable of predictably enhanced cartilage production. Stem Cells 2017;35:2280-2291.


Subject(s)
Chondrogenesis/genetics , Mesenchymal Stem Cells/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Wnt-5a Protein/genetics , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Humans , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Sheep , Tissue Engineering , Wnt-5a Protein/metabolism
6.
Stem Cells Transl Med ; 6(4): 1237-1248, 2017 04.
Article in English | MEDLINE | ID: mdl-28186682

ABSTRACT

Meniscal cartilage tears are common and predispose to osteoarthritis (OA). Most occur in the avascular portion of the meniscus where current repair techniques usually fail. We described previously the use of undifferentiated autologous mesenchymal stem cells (MSCs) seeded onto a collagen scaffold (MSC/collagen-scaffold) to integrate meniscal tissues in vitro. Our objective was to translate this method into a cell therapy for patients with torn meniscus, with the long-term goal of delaying or preventing the onset of OA. After in vitro optimization, we tested an ovine-MSC/collagen-scaffold in a sheep meniscal cartilage tear model with promising results after 13 weeks, although repair was not sustained over 6 months. We then conducted a single center, prospective, open-label first-in-human safety study of patients with an avascular meniscal tear. Autologous MSCs were isolated from an iliac crest bone marrow biopsy, expanded and seeded into the collagen scaffold. The resulting human-MSC/collagen-scaffold implant was placed into the meniscal tear prior to repair with vertical mattress sutures and the patients were followed for 2 years. Five patients were treated and there was significant clinical improvement on repeated measures analysis. Three were asymptomatic at 24 months with no magnetic resonance imaging evidence of recurrent tear and clinical improvement in knee function scores. Two required subsequent meniscectomy due to retear or nonhealing of the meniscal tear at approximately 15 months after implantation. No other adverse events occurred. We conclude that undifferentiated MSCs could provide a safe way to augment avascular meniscal repair in some patients. Registration: EU Clinical Trials Register, 2010-024162-22. Stem Cells Translational Medicine 2017;6:1237-1248.


Subject(s)
Cartilage Diseases/therapy , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Tibial Meniscus Injuries/therapy , Animals , Cell- and Tissue-Based Therapy/methods , Cells, Cultured , Female , Humans , In Vitro Techniques , Menisci, Tibial/cytology , Sheep , Tissue Engineering/methods , Tissue Scaffolds , Wound Healing/physiology
7.
Nat Commun ; 6: 7405, 2015 Jun 17.
Article in English | MEDLINE | ID: mdl-26080734

ABSTRACT

Restricted oxygen diffusion can result in central cell necrosis in engineered tissue, a problem that is exacerbated when engineering large tissue constructs for clinical application. Here we show that pre-treating human mesenchymal stem cells (hMSCs) with synthetic membrane-active myoglobin-polymer-surfactant complexes can provide a reservoir of oxygen capable of alleviating necrosis at the centre of hyaline cartilage. This is achieved through the development of a new cell functionalization methodology based on polymer-surfactant conjugation, which allows the delivery of functional proteins to the hMSC membrane. This new approach circumvents the need for cell surface engineering using protein chimerization or genetic transfection, and we demonstrate that the surface-modified hMSCs retain their ability to proliferate and to undergo multilineage differentiation. The functionalization technology is facile, versatile and non-disruptive, and in addition to tissue oxygenation, it should have far-reaching application in a host of tissue engineering and cell-based therapies.


Subject(s)
Hyaline Cartilage , Mesenchymal Stem Cells/drug effects , Myoglobin/pharmacology , Oxygen/administration & dosage , Tissue Engineering/methods , Escherichia coli , Glycolates/chemistry , Humans , Myoglobin/chemistry
8.
Cartilage ; 6(2 Suppl): 30S-5S, 2015 Apr.
Article in English | MEDLINE | ID: mdl-27340514

ABSTRACT

Chondrogenic progenitor populations, including mesenchymal stem cells, represent promising cell-based transplantation or tissue engineering therapies for the regeneration of damaged cartilage. Osteoarthritis (OA) predominantly affects the elderly and is a leading cause of disability worldwide. Advancing age is a prominent risk factor that is closely associated with the onset and progression of the disease. Understanding the influence that aging and OA have on chondrogenic progenitor cells is important to determine how these processes affect the cellular mechanisms of the cells and their capacity to differentiate into functional chondrocytes for use in therapeutic applications. Here, we review the effect of age- and OA-related changes on the growth kinetics and differentiation potential of chondrogenic progenitor cell populations. Aging differentially influences the proliferative potential of progenitor cells showing reduced growth rates with increased senescence and apoptotic activity over time, while chondrogenesis appears to be independent of donor age. Cartilage tissue affected by OA shows evidence of progenitor populations with some potential for repair, however reports on the proliferative propensity of mesenchymal stem cells and their chondrogenic potential are contradictory. This is likely attributed to the narrow age ranges of samples assessed and deficits in definitively identifying donors with OA versus healthy patients across a wide scope of advancing ages. Further studies that investigate the mechanistic effects of chondrogenic progenitor populations associated with aging and the progression of OA using clearly defined criteria and age-matched control subject groups are crucial to our understanding of the clinical relevance of these cells for use in cartilage repair therapies.

9.
Chem Sci ; 6(11): 6106-6111, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-30090225

ABSTRACT

We present a new approach for the directed delivery of biomolecular payloads to individual cells with high spatial precision. This was accomplished via active sequestration of proteins, oligonucleotides or molecular dyes into coacervate microdroplets, which were then delivered to specific regions of stem cell membranes using a dynamic holographic assembler, resulting in spontaneous coacervate microdroplet-membrane fusion. The facile preparation, high sequestration efficiency and inherent membrane affinity of the microdroplets make this novel "cell paintballing" technology a highly advantageous option for spatially-directed cell functionalization, with potential applications in single cell stimulation, transfection and differentiation.

10.
Stem Cells Dev ; 23(5): 541-54, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24172175

ABSTRACT

Cartilage injuries and osteoarthritis are leading causes of disability in developed countries. The regeneration of damaged articular cartilage using cell transplantation or tissue engineering holds much promise but requires the identification of an appropriate cell source with a high proliferative propensity and consistent chondrogenic capacity. Human fetal mesenchymal stem cells (MSCs) have been isolated from a range of perinatal tissues, including first-trimester bone marrow, and have demonstrated enhanced expansion and differentiation potential. However, their ability to form mature chondrocytes for use in cartilage tissue engineering has not been clearly established. Here, we compare the chondrogenic potential of human MSCs isolated from fetal and adult bone marrow and show distinct differences in their responsiveness to specific growth factors. Transforming growth factor beta 3 (TGFß3) induced chondrogenesis in adult but not fetal MSCs. In contrast, bone morphogenetic protein 2 (BMP2) induced chondrogenesis in fetal but not adult MSCs. When fetal MSCs co-stimulated with BMP2 and TGFß3 were used for cartilage tissue engineering, they generated tissue with type II collagen and proteoglycan content comparable to adult MSCs treated with TGFß3 alone. Investigation of the TGFß/BMP signaling pathway showed that TGFß3 induced phosphorylation of SMAD3 in adult but not fetal MSCs. These findings demonstrate that the initiation of chondrogenesis is modulated by distinct signaling mechanisms in fetal and adult MSCs. This study establishes the feasibility of using fetal MSCs in cartilage repair applications and proposes their potential as an in vitro system for modeling chondrogenic differentiation and skeletal development studies.


Subject(s)
Cell Differentiation/genetics , Chondrogenesis/genetics , Mesenchymal Stem Cells/cytology , Signal Transduction/genetics , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Bone Marrow Cells/cytology , Cartilage/pathology , Cartilage/transplantation , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Tissue Engineering , Transforming Growth Factor beta3
11.
Regen Med ; 9(1): 81-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24351008

ABSTRACT

Those working in the regenerative medicine field currently face numerous regulatory and related challenges. This Perspective captures some of the key ideas of a UK-based working group drawn from academic, clinical and industrial communities and also identifies some key steps that should be taken in the UK and elsewhere to address these challenges.


Subject(s)
Cell- and Tissue-Based Therapy/trends , Genetic Therapy/trends , Government Regulation , Regenerative Medicine/methods , Regenerative Medicine/trends , Clinical Trials as Topic/methods , Genetic Therapy/legislation & jurisprudence , Regenerative Medicine/legislation & jurisprudence , Socioeconomic Factors , United Kingdom
12.
Biomacromolecules ; 14(5): 1287-98, 2013 May 13.
Article in English | MEDLINE | ID: mdl-23534615

ABSTRACT

Biomaterials that can stimulate stem cell differentiation without growth factor supplementation provide potent and cost-effective scaffolds for regenerative medicine. We hypothesize that a scaffold prepared from cellulose and silk blends can direct stem cell chondrogenic fate. We systematically prepared cellulose blends with silk at different compositions using an environmentally benign processing method based on ionic liquids as a common solvent. We tested the effect of blend compositions on the physical properties of the materials as well as on their ability to support mesenchymal stem cell (MSC) growth and chondrogenic differentiation. The stiffness and tensile strength of cellulose was significantly reduced by blending with silk. The characterized materials were tested using MSCs derived from four different patients. Growing MSCs on a specific blend combination of cellulose and silk in a 75:25 ratio significantly upregulated the chondrogenic marker genes SOX9, aggrecan, and type II collagen in the absence of specific growth factors. This chondrogenic effect was neither found with neat cellulose nor the cellulose/silk 50:50 blend composition. No adipogenic or osteogenic differentiation was detected on the blends, suggesting that the cellulose/silk 75:25 blend induced specific stem cell differentiation into the chondrogenic lineage without addition of the soluble growth factor TGF-ß. The cellulose/silk blend we identified can be used both for in vitro tissue engineering and as an implantable device for stimulating endogenous stem cells to initiate cartilage repair.


Subject(s)
Biocompatible Materials/pharmacology , Cellulose/chemistry , Chondrocytes/drug effects , Chondrogenesis/drug effects , Mesenchymal Stem Cells/drug effects , Silk/chemistry , Tissue Engineering/methods , Aggrecans/genetics , Aggrecans/metabolism , Biocompatible Materials/chemistry , Cell Differentiation , Chondrocytes/cytology , Chondrocytes/metabolism , Collagen Type II/genetics , Collagen Type II/metabolism , Gene Expression Regulation/drug effects , Humans , Ionic Liquids , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Tensile Strength , Tissue Scaffolds
14.
Cartilage ; 2(2): 153-72, 2011 Apr.
Article in English | MEDLINE | ID: mdl-26069577

ABSTRACT

Cartilage repair strategies aim to resurface a lesion with osteochondral tissue resembling native cartilage, but a variety of repair tissues are usually observed. Histology is an important structural outcome that could serve as an interim measure of efficacy in randomized controlled clinical studies. The purpose of this article is to propose guidelines for standardized histoprocessing and unbiased evaluation of animal tissues and human biopsies. Methods were compiled from a literature review, and illustrative data were added. In animal models, treatments are usually administered to acute defects created in healthy tissues, and the entire joint can be analyzed at multiple postoperative time points. In human clinical therapy, treatments are applied to developed lesions, and biopsies are obtained, usually from a subset of patients, at a specific time point. In striving to standardize evaluation of structural endpoints in cartilage repair studies, 5 variables should be controlled: 1) location of biopsy/sample section, 2) timing of biopsy/sample recovery, 3) histoprocessing, 4) staining, and 5) blinded evaluation with a proper control group. Histological scores, quantitative histomorphometry of repair tissue thickness, percentage of tissue staining for collagens and glycosaminoglycan, polarized light microscopy for collagen fibril organization, and subchondral bone integration/structure are all relevant outcome measures that can be collected and used to assess the efficacy of novel therapeutics. Standardized histology methods could improve statistical analyses, help interpret and validate noninvasive imaging outcomes, and permit cross-comparison between studies. Currently, there are no suitable substitutes for histology in evaluating repair tissue quality and cartilaginous character.

15.
Stem Cells ; 28(11): 1992-6, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20882533

ABSTRACT

Cartilage is considered to be a simple tissue that should be easy to engineer because it is avascular and contains just one cell type, the chondrocyte. Despite this apparent simplicity, regenerating cartilage in a form that can function effectively after implantation in the joint has proven difficult. This may be because we have not fully appreciated the importance of different structural regions of articular cartilage or of understanding the origins of chondrocytes and how this cell population is maintained in the normal tissue. This review considers what is known about different regions of cartilage and the types of stem cells in articulating joints and emphasizes the potential importance of regeneration of the lamina splendens at the joint surface and calcified cartilage at the junction with bone for long-term survival of regenerated tissue in vivo.


Subject(s)
Cartilage/cytology , Regeneration/physiology , Adult Stem Cells/cytology , Animals , Chondrogenesis/physiology , Humans , Mesenchymal Stem Cells/cytology , Models, Biological
16.
Methods Mol Biol ; 622: 367-78, 2010.
Article in English | MEDLINE | ID: mdl-20135294

ABSTRACT

The biochemistry of collagen makes the assay of its degradation complex. Hidden epitopes are linear amino acid sequences that are not normally available for antibody binding when they are contained within an intact helical structure, but they become exposed once the collagen triple helix has been cleaved and denatured. This chapter describes the use of an antibody raised against such an epitope, combined with selective proteolytic extraction, to assay collagen degradation.


Subject(s)
Biological Assay/methods , Collagen/metabolism , Protein Processing, Post-Translational , Animals , Cattle , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology
17.
Biomaterials ; 31(9): 2583-91, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20053438

ABSTRACT

Injuries to the avascular region of knee meniscal cartilage do not heal spontaneously. To address this problem we have developed a new stem cell/collagen-scaffold implant system in which human adult bone marrow mesenchymal stem cells are seeded onto a biodegradable scaffold that allows controlled delivery of actively dividing cells to the meniscus surface. Sandwich constructs of two white zone ovine meniscus discs with stem cell/collagen-scaffold implant in between were cultured in vitro for 40 days. Histomorphometric analysis revealed superior integration in the stem cell/collagen-scaffold groups compared to the cell-free collagen membrane or untreated controls. The addition of TGF-beta1 to differentiate stem cells to chondrocytes inhibited integration. Biomechanical testing demonstrated a significant 2-fold increase in tensile strength in all constructs using the stem cell/collagen-scaffold compared to control groups after 40 days in culture. Integration was significantly higher when collagen membranes were used that had a more open/spongy structure adjacent to both meniscal cartilage surfaces, whereas a collagen scaffold designed for osteoinduction failed to induce any integration of meniscus. In conclusion, the stem cell/collagen-scaffold implant is a potential therapeutic treatment for the repair of white zone meniscal cartilage tears.


Subject(s)
Cartilage/pathology , Collagen/pharmacology , Implants, Experimental , Menisci, Tibial/pathology , Mesenchymal Stem Cells/cytology , Tissue Scaffolds/chemistry , Wound Healing/drug effects , Animals , Cartilage/drug effects , Cattle , Cell Differentiation/drug effects , Chemotaxis/drug effects , Humans , Mechanical Phenomena/drug effects , Membranes, Artificial , Menisci, Tibial/drug effects , Mesenchymal Stem Cells/drug effects , Sheep
18.
Biomaterials ; 30(29): 5260-9, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19647867

ABSTRACT

Cell and tissue engineering are now being translated into clinical organ replacement, offering alternatives to fight morbidity, organ shortages and ethico-social problems associated with allotransplantation. Central to the recent first successful use of stem cells to create an organ replacement in man was our development of a bioreactor environment. Critical design features were the abilities to drive the growth of two different cell types, to support 3D maturation, to maintain biomechanical and biological properties and to provide appropriate hydrodynamic stimuli and adequate mass transport. An analytical model was developed and applied to predict oxygen profiles in the bioreactor-cultured organ construct and in the culture media, comparing representative culture configurations and operating conditions. Autologous respiratory epithelial cells and mesenchymal stem cells (BMSCs, then differentiated into chondrocytes) were isolated, characterized and expanded. Both cell types were seeded and cultured onto a decellularized human donor tracheal matrix within the bioreactor. One year post-operatively, graft and patient are healthy, and biopsies confirm angiogenesis, viable epithelial cells and chondrocytes. Our rotating double-chamber bioreactor permits the efficient repopulation of a decellularized human matrix, a concept that can be applied clinically, as demonstrated by the successful tracheal transplantation.


Subject(s)
Bioartificial Organs , Bioreactors , Epithelial Cells/transplantation , Mesenchymal Stem Cell Transplantation/methods , Organ Culture Techniques/instrumentation , Tissue Engineering/instrumentation , Trachea/growth & development , Trachea/transplantation , Epithelial Cells/cytology , Epithelial Cells/physiology , Equipment Design , Equipment Failure Analysis , Humans , Rotation , Treatment Outcome
19.
Biomaterials ; 30(26): 4277-86, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19539365

ABSTRACT

The integration of implanted cartilage is a major challenge for the success of tissue engineering protocols. We hypothesize that in order for effective cartilage integration to take place, matrix-free chondrocytes must be induced to migrate between the two tissue surfaces. A chondrocyte/collagen-scaffold implant system was developed as a method of delivering dividing cells at the interface between two cartilage surfaces. Chondrocytes were isolated from bovine nasal septum and seeded onto both surfaces of a collagen membrane to create the chondrocyte/collagen-scaffold implant. A model of two cartilage discs and the chondrocyte/collagen-scaffold sandwiched in between was used to effect integration in vitro. The resulting tissue was analysed histologically and biomechanically. The cartilage-implant-cartilage sandwich appeared macroscopically as one continuous piece of tissue at the end of 40 day cultures. Histological analysis showed tissue continuum across the cartilage-scaffold interface. The integration was dependent on both cells and scaffold. Fluorescent labeling of implanted chondrocytes demonstrated that these cells invade the surrounding mature tissue and drive a remodelling of the extracellular matrix. Using cell-free scaffolds we also demonstrated that some chondrocytes migrated from the natural cartilage into the collagen scaffold. Quantification of integration levels using a histomorphometric repair index showed that the chondrocyte/collagen-scaffold implant achieved the highest repair index compared to controls, reflected functionally through increased tensile strength. In conclusion, cartilage integration can be achieved using a chondrocyte/collagen-scaffold implant that permits controlled delivery of chondrocytes to both host and graft mature cartilage tissues. This approach has the potential to be used therapeutically for implantation of engineered tissue.


Subject(s)
Cartilage/metabolism , Chondrocytes/metabolism , Collagen/metabolism , Implants, Experimental , Osseointegration , Tissue Scaffolds , Animals , Cartilage/cytology , Cattle , Cell Movement , Chondrocytes/cytology , Extracellular Matrix/metabolism , Tensile Strength
20.
Lancet ; 372(9655): 2023-30, 2008 12 13.
Article in English | MEDLINE | ID: mdl-19022496

ABSTRACT

BACKGROUND: The loss of a normal airway is devastating. Attempts to replace large airways have met with serious problems. Prerequisites for a tissue-engineered replacement are a suitable matrix, cells, ideal mechanical properties, and the absence of antigenicity. We aimed to bioengineer tubular tracheal matrices, using a tissue-engineering protocol, and to assess the application of this technology in a patient with end-stage airway disease. METHODS: We removed cells and MHC antigens from a human donor trachea, which was then readily colonised by epithelial cells and mesenchymal stem-cell-derived chondrocytes that had been cultured from cells taken from the recipient (a 30-year old woman with end-stage bronchomalacia). This graft was then used to replace the recipient's left main bronchus. FINDINGS: The graft immediately provided the recipient with a functional airway, improved her quality of life, and had a normal appearance and mechanical properties at 4 months. The patient had no anti-donor antibodies and was not on immunosuppressive drugs. INTERPRETATION: The results show that we can produce a cellular, tissue-engineered airway with mechanical properties that allow normal functioning, and which is free from the risks of rejection. The findings suggest that autologous cells combined with appropriate biomaterials might provide successful treatment for patients with serious clinical disorders.


Subject(s)
Bronchomalacia/physiopathology , Chondrocytes/cytology , Tissue Engineering/methods , Trachea/transplantation , Adult , Bronchomalacia/therapy , Cadaver , Female , Humans , Postoperative Period , Respiratory Function Tests , Trachea/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...