Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
Cytotherapy ; 25(5): 548-558, 2023 05.
Article En | MEDLINE | ID: mdl-36894437

The development of medicinal products often continues throughout the different phases of a clinical study and may require challenging changes in raw and starting materials at later stages. Comparability between the product properties pre- and post-change thus needs to be ensured. Here, we describe and validate the regulatory compliant change of a raw material using the example of a nasal chondrocyte tissue-engineered cartilage (N-TEC) product, initially developed for treatment of confined knee cartilage lesions. Scaling up the size of N-TEC as required for the treatment of larger osteoarthritis defects required the substitution of autologous serum with a clinical-grade human platelet lysate (hPL) to achieve greater cell numbers necessary for the manufacturing of larger size grafts. A risk-based approach was performed to fulfill regulatory requirements and demonstrate comparability of the products manufactured with the standard process (autologous serum) already applied in clinical indications and the modified process (hPL). Critical attributes with regard to quality, purity, efficacy, safety and stability of the product as well as associated test methods and acceptance criteria were defined. Results showed that hPL added during the expansion phase of nasal chondrocytes enhances proliferation rate, population doublings and cell numbers at passage 2 without promoting the overgrowth of potentially contaminant perichondrial cells. N-TEC generated with the modified versus standard process contained similar content of DNA and cartilaginous matrix proteins with even greater expression levels of chondrogenic genes. The increased risk for tumorigenicity potentially associated with the use of hPL was assessed through karyotyping of chondrocytes at passage 4, revealing no chromosomal changes. Moreover, the shelf-life of N-TEC established for the standard process could be confirmed with the modified process. In conclusion, we demonstrated the introduction of hPL in the manufacturing process of a tissue engineered product, already used in a late-stage clinical trial. Based on this study, the national competent authorities in Switzerland and Germany accepted the modified process which is now applied for ongoing clinical tests of N-TEC. The described activities can thus be taken as a paradigm for successful and regulatory compliant demonstration of comparability in advanced therapy medicinal products manufacturing.


Chondrocytes , Tissue Engineering , Humans , Karyotyping , Knee Joint
2.
Front Oncol ; 11: 775136, 2021.
Article En | MEDLINE | ID: mdl-34938659

The reconstruction of complex midface defects is a challenging clinical scenario considering the high anatomical, functional, and aesthetic requirements. In this study, we proposed a surgical treatment to achieve improved oral rehabilitation and anatomical and functional reconstruction of a complex defect of the maxilla with a vascularized, engineered composite graft. The patient was a 39-year-old female, postoperative after left hemimaxillectomy for ameloblastic carcinoma in 2010 and tumor-free at the 5-year oncological follow-up. The left hemimaxillary defect was restored in a two-step approach. First, a composite graft was ectopically engineered using autologous stromal vascular fraction (SVF) cells seeded on an allogenic devitalized bone matrix. The resulting construct was further loaded with bone morphogenic protein-2 (BMP-2), wrapped within the latissimus dorsi muscle, and pedicled with an arteriovenous (AV) bundle. Subsequently, the prefabricated graft was orthotopically transferred into the defect site and revascularized through microvascular surgical techniques. The prefabricated graft contained vascularized bone tissue embedded within muscular tissue. Despite unexpected resorption, its orthotopic transfer enabled restoration of the orbital floor, separation of the oral and nasal cavities, and midface symmetry and allowed the patient to return to normal diet as well as to restore normal speech and swallowing function. These results remained stable for the entire follow-up period of 2 years. This clinical case demonstrates the safety and the feasibility of composite graft engineering for the treatment of complex maxillary defects. As compared to the current gold standard of autologous tissue transfer, this patient's benefits included decreased donor site morbidity and improved oral rehabilitation. Bone resorption of the construct at the ectopic prefabrication site still needs to be further addressed to preserve the designed graft size and shape.

3.
Front Med (Lausanne) ; 8: 712917, 2021.
Article En | MEDLINE | ID: mdl-34485343

Advanced Therapy Medicinal Products (ATMP) provide promising treatment options particularly for unmet clinical needs, such as progressive and chronic diseases where currently no satisfying treatment exists. Especially from the ATMP subclass of Tissue Engineered Products (TEPs), only a few have yet been translated from an academic setting to clinic and beyond. A reason for low numbers of TEPs in current clinical trials and one main key hurdle for TEPs is the cost and labor-intensive manufacturing process. Manual production steps require experienced personnel, are challenging to standardize and to scale up. Automated manufacturing has the potential to overcome these challenges, toward an increasing cost-effectiveness. One major obstacle for automation is the control and risk prevention of cross contaminations, especially when handling parallel production lines of different patient material. These critical steps necessitate validated effective and efficient cleaning procedures in an automated system. In this perspective, possible technologies, concepts and solutions to existing ATMP manufacturing hurdles are discussed on the example of a late clinical phase II trial TEP. In compliance to Good Manufacturing Practice (GMP) guidelines, we propose a dual arm robot based isolator approach. Our novel concept enables complete process automation for adherent cell culture, and the translation of all manual process steps with standard laboratory equipment. Moreover, we discuss novel solutions for automated cleaning, without the need for human intervention. Consequently, our automation concept offers the unique chance to scale up production while becoming more cost-effective, which will ultimately increase TEP availability to a broader number of patients.

4.
Clin Hemorheol Microcirc ; 74(1): 67-78, 2020.
Article En | MEDLINE | ID: mdl-31743993

OBJECTIVE: Implantation of autologous chondrocytes for cartilage repair requires harvesting of undamaged cartilage, implying an additional joint arthroscopy surgery and further damage to the articular surface. As alternative possible cell sources, in this study we assessed the proliferation and chondrogenic capacity of debrided Knee Chondrocytes (dKC) and Nasal Chondrocytes (NC) collected from the same patients. METHODS: Matched NC and dKC pairs from 13 patients enrolled in two clinical studies (NCT01605201 and NCT026739059) were expanded in monolayer and then chondro-differentiated in 3D collagenous scaffolds in medium with or without Transforming Growth Factor beta 1 (TGFß1). Cell proliferation and amount of cartilage matrix production by these two cell types were assessed. RESULTS: dKC exhibited an inferior proliferation rate than NC, and a lower capacity to chondro-differentiate. Resulting dKC-grafts contained lower amounts of cartilage specific matrix components glycosaminoglycans and type II collagen. The cartilage forming capacity of dKC did not significantly correlate with specific clinical parameters and was only partially improved by medium supplemention with TGFß1. CONCLUSIONS: dKC exhibit a reproducibly poor capacity to engineer cartilage grafts. Our in vitro data suggest that NC would be a better suitable cell source for the generation of autologous cartilage grafts.


Cartilage, Articular/physiopathology , Chondrocytes/metabolism , Knee Joint/physiopathology , Nose/physiopathology , Tissue Engineering/methods , Adult , Cell Differentiation , Cell Proliferation , Cells, Cultured , Female , Humans , Male , Middle Aged
5.
Swiss Med Wkly ; 149: w20032, 2019 Mar 25.
Article En | MEDLINE | ID: mdl-30950502

The effects of oncological treatment, congenital anomalies, traumatic injuries and post-infection damage critically require sufficient amounts of tissue for structural and functional surgical reconstructions. The patient’s own body is typically the gold standard source of transplant material, but in children autologous tissue is available only in small quantities and with severe morbidity at donor sites. Engineering of tissue grafts starting from a small amount of autologous material, combined with suitable surgical manipulation of the recipient site, is expected to enhance child and adolescent health, and to offer functional restoration for long-term wellbeing. Moreover, engineered tissues based on patient-derived cells represent invaluable models to investigate mechanisms of disease and to develop/test novel therapeutic approaches. In view of these great opportunities, here we introduce the currently limited successful implementation of tissue engineering in paediatric settings and discuss the open challenges in the field. A particular focus is on the specific needs and envisioned strategies in the areas of bone and osteochondral regeneration in children.


Cell- and Tissue-Based Therapy/methods , Pediatrics/methods , Regenerative Medicine/methods , Tissue Engineering/methods , Adolescent , Bone Regeneration , Child , Female , Humans , Male , Transplantation, Autologous
6.
Lancet ; 388(10055): 1985-1994, 2016 Oct 22.
Article En | MEDLINE | ID: mdl-27789021

BACKGROUND: Articular cartilage injuries have poor repair capacity, leading to progressive joint damage, and cannot be restored predictably by either conventional treatments or advanced therapies based on implantation of articular chondrocytes. Compared with articular chondrocytes, chondrocytes derived from the nasal septum have superior and more reproducible capacity to generate hyaline-like cartilage tissues, with the plasticity to adapt to a joint environment. We aimed to assess whether engineered autologous nasal chondrocyte-based cartilage grafts allow safe and functional restoration of knee cartilage defects. METHODS: In a first-in-human trial, ten patients with symptomatic, post-traumatic, full-thickness cartilage lesions (2-6 cm2) on the femoral condyle or trochlea were treated at University Hospital Basel in Switzerland. Chondrocytes isolated from a 6 mm nasal septum biopsy specimen were expanded and cultured onto collagen membranes to engineer cartilage grafts (30 × 40 × 2 mm). The engineered tissues were implanted into the femoral defects via mini-arthrotomy and assessed up to 24 months after surgery. Primary outcomes were feasibility and safety of the procedure. Secondary outcomes included self-assessed clinical scores and MRI-based estimation of morphological and compositional quality of the repair tissue. This study is registered with ClinicalTrials.gov, number NCT01605201. The study is ongoing, with an approved extension to 25 patients. FINDINGS: For every patient, it was feasible to manufacture cartilaginous grafts with nasal chondrocytes embedded in an extracellular matrix rich in glycosaminoglycan and type II collagen. Engineered tissues were stable through handling with forceps and could be secured in the injured joints. No adverse reactions were recorded and self-assessed clinical scores for pain, knee function, and quality of life were improved significantly from before surgery to 24 months after surgery. Radiological assessments indicated variable degrees of defect filling and development of repair tissue approaching the composition of native cartilage. INTERPRETATION: Hyaline-like cartilage tissues, engineered from autologous nasal chondrocytes, can be used clinically for repair of articular cartilage defects in the knee. Future studies are warranted to assess efficacy in large controlled trials and to investigate an extension of indications to early degenerative states or to other joints. FUNDING: Deutsche Arthrose-Hilfe.


Cartilage, Articular/surgery , Chondrocytes/transplantation , Knee Joint/surgery , Nasal Septum/cytology , Tissue Engineering , Transplants , Adult , Cartilage, Articular/injuries , Cartilage, Articular/pathology , Evidence-Based Medicine , Feasibility Studies , Female , Follow-Up Studies , Humans , Knee Joint/diagnostic imaging , Knee Joint/physiopathology , Male , Middle Aged , Minimally Invasive Surgical Procedures , Pain/etiology , Quality of Life , Recovery of Function , Self Report , Switzerland , Tissue Scaffolds , Transplantation, Autologous , Treatment Outcome
7.
Stem Cells ; 34(12): 2956-2966, 2016 12.
Article En | MEDLINE | ID: mdl-27538760

Stromal Vascular Fraction (SVF) cells freshly isolated from adipose tissue include osteogenic- and vascular-progenitors, yet their relevance in bone fracture healing is currently unknown. Here, we investigated whether human SVF cells directly contribute to the repair of experimental fractures in nude rats, and explored the feasibility/safety of their clinical use for augmentation of upper arm fractures in elderly individuals. Human SVF cells were loaded onto ceramic granules within fibrin gel and implanted in critical nude rat femoral fractures after locking-plate osteosynthesis, with cell-free grafts as control. After 8 weeks, only SVF-treated fractures did not fail mechanically and displayed formation of ossicles at the repair site, with vascular and bone structures formed by human cells. The same materials combined with autologous SVF cells were then used to treat low-energy proximal humeral fractures in 8 patients (64-84 years old) along with standard open reduction and internal fixation. Graft manufacturing and implantation were compatible with intraoperative settings and led to no adverse reactions, thereby verifying feasibility/safety. Biopsies of the repair tissue after up to 12 months, upon plate revision or removal, demonstrated formation of bone ossicles, structurally disconnected and morphologically distinct from osteoconducted bone, suggesting the osteogenic nature of implanted SVF cells. We demonstrate that SVF cells, without expansion or exogenous priming, can spontaneously form bone tissue and vessel structures within a fracture-microenvironment. The gained clinical insights into the biological functionality of the grafts, combined with their facile, intra-operative manufacturing modality, warrant further tests of effectiveness in larger, controlled trials. Stem Cells 2016;34:2956-2966.


Fractures, Bone/pathology , Stem Cell Transplantation , Stem Cells/cytology , Aged , Aged, 80 and over , Animals , Demography , Disease Models, Animal , Female , Femur/diagnostic imaging , Femur/pathology , Follow-Up Studies , Fractures, Bone/diagnostic imaging , Fractures, Bone/therapy , Humans , Immunohistochemistry , Male , Middle Aged , Osteogenesis , Pain Measurement , Rats , Stromal Cells/transplantation
8.
Lancet ; 384(9940): 337-46, 2014 Jul 26.
Article En | MEDLINE | ID: mdl-24726477

BACKGROUND: Autologous native cartilage from the nasal septum, ear, or rib is the standard material for surgical reconstruction of the nasal alar lobule after two-layer excision of non-melanoma skin cancer. We assessed whether engineered autologous cartilage grafts allow safe and functional alar lobule restoration. METHODS: In a first-in-human trial, we recruited five patients at the University Hospital Basel (Basel, Switzerland). To be eligible, patients had to be aged at least 18 years and have a two-layer defect (≥50% size of alar subunit) after excision of non-melanoma skin cancer on the alar lobule. Chondrocytes (isolated from a 6 mm cartilage biopsy sample from the nasal septum harvested under local anaesthesia during collection of tumour biopsy sample) were expanded, seeded, and cultured with autologous serum onto collagen type I and type III membranes in the course of 4 weeks. The resulting engineered cartilage grafts (25 mm × 25 mm × 2 mm) were shaped intra-operatively and implanted after tumour excision under paramedian forehead or nasolabial flaps, as in standard reconstruction with native cartilage. During flap refinement after 6 months, we took biopsy samples of repair tissues and histologically analysed them. The primary outcomes were safety and feasibility of the procedure, assessed 12 months after reconstruction. At least 1 year after implantation, when reconstruction is typically stabilised, we assessed patient satisfaction and functional outcomes (alar cutaneous sensibility, structural stability, and respiratory flow rate). FINDINGS: Between Dec 13, 2010, and Feb 6, 2012, we enrolled two women and three men aged 76-88 years. All engineered grafts contained a mixed hyaline and fibrous cartilage matrix. 6 months after implantation, reconstructed tissues displayed fibromuscular fatty structures typical of the alar lobule. After 1 year, all patients were satisfied with the aesthetic and functional outcomes and no adverse events had been recorded. Cutaneous sensibility and structural stability of the reconstructed area were clinically satisfactory, with adequate respiratory function. INTERPRETATION: Autologous nasal cartilage tissues can be engineered and clinically used for functional restoration of alar lobules. Engineered cartilage should now be assessed for other challenging facial reconstructions. FUNDING: Foundation of the Department of Surgery, University Hospital Basel; and Krebsliga beider Basel.


Nasal Cartilages/surgery , Nose Neoplasms/surgery , Skin Neoplasms/surgery , Tissue Engineering/methods , Aged , Aged, 80 and over , Chondrocytes/metabolism , Female , Humans , Male , Middle Aged , Plastic Surgery Procedures/methods
9.
Biomaterials ; 34(33): 8161-71, 2013 Nov.
Article En | MEDLINE | ID: mdl-23896003

We have investigated monocyte and T cell responsiveness to silk based biomaterials of different physico-chemical characteristics. Here we report that untransformed CD14+ human monocytes respond to overnight exposure to silk fibroin-based biomaterials in tridimensional form by IL-1ß and IL-6, but not IL-10 gene expression and protein production. In contrast, fibroin based materials in bidimensional form are unable to stimulate monocyte responsiveness. The elicitation of these effects critically requires contact between biomaterials and responding cells, is not sustained and becomes undetectable in longer term cultures. We also observed that NF-κß and p38 MAP kinase play key roles in monocyte activation by silk-based biomaterials. On the other hand, fibroin based materials, irrespective of their physico-chemical characteristics appeared to be unable to induce the activation of peripheral blood T cells from healthy donors, as evaluated by the expression of activation markers and IFN-γ gene.


Adaptive Immunity/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Immunity, Innate/drug effects , Silk/chemistry , Animals , Cells, Cultured , Cytokines/metabolism , Humans , Interleukin-10/metabolism , Interleukin-6/metabolism , Microscopy, Atomic Force , Monocytes/drug effects , Monocytes/metabolism , NF-kappa B/metabolism , Spectroscopy, Fourier Transform Infrared
10.
Facial Plast Surg ; 29(2): 99-105, 2013 Apr.
Article En | MEDLINE | ID: mdl-23564241

Since the late 1960s, surgeons and scientists envisioned use of tissue engineering to provide an alternative treatment for tissue and organ damage by combining biological and synthetic components in such a way that a long-lasting repair was established. In addition to the treatment, the patient would also benefit from reduced donor site morbidity and operation time as compared with the standard procedures. Tremendous efforts in basic research have been done since the late 1960s to better understand chondrocyte biology and cartilage maturation and to fulfill the growing need for tissue-engineered cartilage in reconstructive, trauma, and orthopedic surgery. Starting from the first successful generation of engineered cartilaginous tissue, scientists strived to improve the properties of the cartilaginous constructs by characterizing different cell sources, modifying the environmental factors influencing cell expansion and differentiation and applying physical stimuli to modulate the mechanical properties of the construct. All these efforts have finally led to a clinical phase I trial to show the safety and feasibility of using tissue-engineered cartilage in reconstructive facial surgery. However, to bring tissue engineering into routine clinical applications and commercialize tissue-engineered grafts, further research is necessary to achieve a cost-effective, standardized, safe, and regulatory compliant process.


Cartilage , Clinical Trials, Phase I as Topic , Tissue Engineering , Animals , Bioreactors , Cell Culture Techniques , Cell Dedifferentiation , Chondrocytes/cytology , Clinical Trials, Phase I as Topic/economics , Clinical Trials, Phase I as Topic/legislation & jurisprudence , Humans , Switzerland , Tissue Scaffolds
11.
Acta Biomater ; 8(9): 3313-25, 2012 Sep.
Article En | MEDLINE | ID: mdl-22641105

A novel design of silk-based scaffold is developed using a custom-made winding machine, with fiber alignment resembling the anatomical criss-cross lamellar fibrous orientation features of the annulus fibrosus of the intervertebral disc. Crosslinking of silk fibroin fibers with chondroitin sulphate (CS) was introduced to impart superior biological functionality. The scaffolds, with or without CS, instructed alignment of expanded human chondrocytes and of the deposited extracellular matrix while supporting their chondrogenic redifferentiation. The presence of CS crosslinking could not induce statistically significant changes in the measured collagen or glycosaminoglycan content, but resulted in an increased construct stiffness. By offering the combined effect of cell/matrix alignment and chondrogenic support, the silk fibroin scaffolds developed with precise fiber orientation in lamellar form represent a suitable substrate for tissue engineering of the annulus fibrosus part of the intervertebral disc.


Cartilage/chemistry , Silk , Tissue Engineering , Cells, Cultured , Humans , Microscopy, Atomic Force , Microscopy, Electron, Transmission , Models, Molecular , Spectroscopy, Fourier Transform Infrared
12.
Tissue Eng Part A ; 18(3-4): 362-72, 2012 Feb.
Article En | MEDLINE | ID: mdl-21902467

Previous studies showed that human nasal chondrocytes (HNC) exhibit higher proliferation and chondrogenic capacity as compared to human articular chondrocytes (HAC). To consider HNC as a relevant alternative cell source for the repair of articular cartilage defects it is necessary to test how these cells react when exposed to environmental factors typical of an injured joint. We thus aimed this study at investigating the responses of HNC and HAC to exposure to interleukin (IL)-1ß and low oxygen. For this purpose HAC and HNC harvested from the same donors (N=5) were expanded in vitro and then cultured in pellets or collagen-based scaffolds at standard (19%) or low oxygen (5%) conditions. Resulting tissues were analyzed after a short (3 days) exposure to IL-1ß, mimicking the initially inflammatory implantation site, or following a recovery time (1 or 2 weeks for pellets and scaffolds, respectively). After IL-1ß treatment, constructs generated by both HAC and HNC displayed a transient loss of GAG (up to 21.8% and 36.8%, respectively) and, consistently, an increased production of metalloproteases (MMP)-1 and -13. Collagen type II and the cryptic fragment of aggrecan (DIPEN), both evaluated immunohistochemically, displayed a trend consistent with GAG and MMPs production. HNC-based constructs exhibited a more efficient recovery upon IL-1ß withdrawal, resulting in a higher accumulation of GAG (up to 2.6-fold) compared to the corresponding HAC-based tissues. On the other hand, HAC displayed a positive response to low oxygen culture, while HNC were only slightly affected by oxygen percentage. Collectively, under the conditions tested mimicking the postsurgery articular environment, HNC retained a tissue-forming capacity, similar or even better than HAC. These results represent a step forward in validating HNC as a cell source for cartilage tissue engineering strategies.


Cartilage, Articular/cytology , Chondrocytes/cytology , Chondrocytes/drug effects , Interleukin-1beta/pharmacology , Nose/cytology , Oxygen/pharmacology , Tissue Engineering/methods , Adult , Aged , Aged, 80 and over , Cartilage Oligomeric Matrix Protein , Cartilage, Articular/drug effects , Cartilage, Articular/physiology , Cells, Cultured , Chondrocytes/enzymology , Chondrocytes/metabolism , Collagen Type II/metabolism , Epitopes/metabolism , Extracellular Matrix Proteins/metabolism , Female , Glycoproteins/metabolism , Glycosaminoglycans/metabolism , Humans , Male , Matrilin Proteins , Matrix Metalloproteinases/metabolism , Middle Aged , Tissue Scaffolds/chemistry
13.
Tissue Eng Part C Methods ; 16(1): 11-21, 2010 Feb.
Article En | MEDLINE | ID: mdl-19327004

We investigated whether human articular chondrocytes can be labeled efficiently and for long-term with a green fluorescent protein (GFP) lentivirus and whether the viral transduction would influence cell proliferation and tissue-forming capacity. The method was then applied to track goat articular chondrocytes after autologous implantation in cartilage defects. Expression of GFP in transduced chondrocytes was detected cytofluorimetrically and immunohistochemically. Chondrogenic capacity of chondrocytes was assessed by Safranin-O staining, immunostaining for type II collagen, and glycosaminoglycan content. Human articular chondrocytes were efficiently transduced with GFP lentivirus (73.4 +/- 0.5% at passage 1) and maintained the expression of GFP up to 22 weeks of in vitro culture after transduction. Upon implantation in nude mice, 12 weeks after transduction, the percentage of labeled cells (73.6 +/- 3.3%) was similar to the initial one. Importantly, viral transduction of chondrocytes did not affect the cell proliferation rate, chondrogenic differentiation, or tissue-forming capacity, either in vitro or in vivo. Goat articular chondrocytes were also efficiently transduced with GFP lentivirus (78.3 +/- 3.2%) and maintained the expression of GFP in the reparative tissue after orthotopic implantation. This study demonstrates the feasibility of efficient and relatively long-term labeling of human chondrocytes for co-culture on integration studies, and indicates the potential of this stable labeling technique for tracking animal chondrocytes for in cartilage repair studies.


Chondrocytes/metabolism , Green Fluorescent Proteins/metabolism , Lentivirus/genetics , Animals , Cell Differentiation , Cell Proliferation , Chondrocytes/cytology , Coculture Techniques , Collagen Type II/metabolism , Glycosaminoglycans/metabolism , Goats , Humans , Immunohistochemistry/methods , Mice , Mice, Nude , Phenazines/pharmacology , Reproducibility of Results
14.
Nat Nanotechnol ; 4(3): 186-92, 2009 Mar.
Article En | MEDLINE | ID: mdl-19265849

The pathological changes in osteoarthritis--a degenerative joint disease prevalent among older people--start at the molecular scale and spread to the higher levels of the architecture of articular cartilage to cause progressive and irreversible structural and functional damage. At present, there are no treatments to cure or attenuate the degradation of cartilage. Early detection and the ability to monitor the progression of osteoarthritis are therefore important for developing effective therapies. Here, we show that indentation-type atomic force microscopy can monitor age-related morphological and biomechanical changes in the hips of normal and osteoarthritic mice. Early damage in the cartilage of osteoarthritic patients undergoing hip or knee replacements could similarly be detected using this method. Changes due to aging and osteoarthritis are clearly depicted at the nanometre scale well before morphological changes can be observed using current diagnostic methods. Indentation-type atomic force microscopy may potentially be developed into a minimally invasive arthroscopic tool to diagnose the early onset of osteoarthritis in situ.


Aging/pathology , Microscopy, Atomic Force , Osteoarthritis/diagnosis , Osteoarthritis/pathology , Animals , Biopsy , Cartilage, Articular/pathology , Cartilage, Articular/ultrastructure , Collagen Type IX/deficiency , Early Diagnosis , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Surface Properties
15.
Matrix Biol ; 27(6): 526-34, 2008 Jul.
Article En | MEDLINE | ID: mdl-18534835

Lumican is a glycoprotein that is found in the extracellular matrix of many connective tissues, including cartilage. It is a member of the small leucine-rich repeat proteoglycans family and along with two others, decorin and fibromodulin, has the capacity to bind to fibrillar collagens and limit their growth. Cartilage tissue engineering provides a potential method for the production of three-dimensional tissue for implantation into eroded joints. Many studies have demonstrated the growth of cartilage in vitro. However in all cases, biochemical analysis of the tissue revealed a significant deficit in the collagen content. We have now tested the hypothesis that the reduced collagen accumulation in engineered cartilage is a result of over-expression of decorin, fibromodulin or lumican. We have found that the lumican gene and protein are both over-expressed in engineered compared to natural cartilage whereas this is not the case for decorin or fibromodulin. Using a small hairpin lumican antisense sequence we were able to knockdown the lumican gene and protein expression in chondrocytes being used for tissue engineering. This resulted in increased accumulation of type II collagen (the major collagen of cartilage) whilst there was no significant alteration in the proteoglycan content. Furthermore, the antisense knockdown of lumican resulted in an increase in the average collagen fibril diameter measured by transmission electron microscopy. These results suggest that lumican plays a pivotal role in the development of tissue engineered cartilage and that regulation of this protein may be important for the production of high-quality implants.


Cartilage , Chondroitin Sulfate Proteoglycans/metabolism , Collagen/metabolism , Keratan Sulfate/metabolism , Tissue Engineering , Animals , Cartilage/cytology , Cartilage/metabolism , Cattle , Chondroitin Sulfate Proteoglycans/genetics , Collagen/ultrastructure , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Gene Deletion , Humans , Keratan Sulfate/genetics , Lumican , Mice , Proteoglycans/genetics , Proteoglycans/metabolism , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Random Allocation
16.
J Biomech ; 40(4): 750-65, 2007.
Article En | MEDLINE | ID: mdl-16730354

Osteochondral defects (i.e., defects which affect both the articular cartilage and underlying subchondral bone) are often associated with mechanical instability of the joint, and therefore with the risk of inducing osteoarthritic degenerative changes. Current surgical limits in the treatment of complex joint lesions could be overcome by grafting osteochondral composite tissues, engineered by combining the patient's own cells with three-dimensional (3D) porous biomaterials of pre-defined size and shape. Various strategies have been reported for the engineering of osteochondral composites, which result from the use of one or more cell types cultured into single-component or composite scaffolds in a broad spectrum of compositions and biomechanical properties. The variety of concepts and models proposed by different groups for the generation of osteochondral grafts reflects that understanding of the requirements to restore a normal joint function is still poor. In order to introduce the use of engineered osteochondral composites in the routine clinical practice, it will be necessary to comprehensively address a number of critical issues, including those related to the size and shape of the graft to be generated, the cell type(s) and properties of the scaffold(s) to be used, the potential physical conditioning to be applied, the degree of functionality required, and the strategy for a cost-effective manufacturing. The progress made in material science, cell biology, mechanobiology and bioreactor technology will be key to support advances in this challenging field.


Bioartificial Organs/trends , Cartilage, Articular , Chondrogenesis/physiology , Tissue Engineering/methods , Biocompatible Materials , Cell Culture Techniques , Chondrocytes/cytology , Osteoblasts/cytology , Tissue Engineering/trends , Transplants
17.
Ann Surg ; 244(6): 978-85; discussion 985, 2006 Dec.
Article En | MEDLINE | ID: mdl-17122623

OBJECTIVE: To investigate if precultivation of human engineered nasal cartilage grafts of clinically relevant size would increase the suture retention strength at implantation and the tensile and bending stiffness 2 weeks after implantation. SUMMARY BACKGROUND INFORMATION: To be used for reconstruction of nasal cartilage defects, engineered grafts need to be reliably sutured at implantation and resist to bending/tension forces about 2 weeks after surgery, when fixation is typically removed. METHODS: Nasal septum chondrocytes from 4 donors were expanded for 2 passages and statically loaded on 15 x 5 x 2-mm size nonwoven meshes of esterified hyaluronan (Hyaff-11). Constructs were implanted for 2 weeks in nude mice between muscle fascia and subcutaneous tissue either directly after cell seeding or after 2 or 4 weeks of preculture in chondrogenic medium. Engineered tissues and native nasal cartilage were assessed histologically, biochemically, and biomechanically. RESULTS: Engineered constructs reproducibly developed with culture time into cartilaginous tissues with increasing content of glycosaminoglycans and collagen type II. Suture retention strength was significantly higher (3.6 +/- 2.2-fold) in 2-week precultured constructs than in freshly seeded meshes. Following in vivo implantation, tissues further developed and maintained the original scaffold size and shape. The bending stiffness was significantly higher (1.8 +/- 0.8-fold) if constructs were precultured for 2 weeks than if they were directly implanted, whereas tensile stiffness was close to native cartilage in all groups. CONCLUSION: In our experimental setup, preculture for 2 weeks was necessary to engineer nasal cartilage grafts with enhanced mechanical properties relevant for clinical use in facial reconstructive surgery.


Chondrocytes/physiology , Nasal Septum/cytology , Rhinoplasty , Tissue Culture Techniques/methods , Tissue Engineering , Adult , Animals , Humans , Hyaluronic Acid/analogs & derivatives , Mice , Middle Aged , Pliability , Suture Techniques , Tensile Strength
18.
Biomacromolecules ; 7(11): 3012-8, 2006 Nov.
Article En | MEDLINE | ID: mdl-17096526

Therapeutic strategies based on cell and tissue engineering can be advanced by developing material substrates that effectively interrogate the biological compartment, with or without the complimentary local release of growth factors. Poly(ether ester) segmented copolymers were engineered as model material systems to elucidate the interfacial molecular events that govern the function of adhered cells. Surface chemistry was modulated by varying poly(ethylene glycol) (PEG) length and mole fraction with poly(butylene terephthalate) (PBT), leading to differential competitive protein adsorption of fibronectin and vitronectin from serum and consequently to different cell attachment modes. Adhesion within the hydrogel-like milieu of longer surface PEG was mediated via binding to the CD44 transmembrane receptor, rather than the RGD-integrin mechanism, whereas greater substrate-bound fibronectin resulted in cell adhesion via integrins. These adhesion modalities differentially impacted morphological cell phenotype (spread or spheroid) and the subsequent expression of mRNA transcripts (collagen types II, I) characteristic of phenotypically differentiated or dedifferentiated chondrocytes, respectively. These results demonstrate that materials can be designed to directly elicit the membrane bound receptor apparatus desired for downstream cellular response, without requiring exogenous biological growth factors to enable differentiated potential.


Chondrocytes/cytology , Polymers/chemistry , Tissue Engineering , Blotting, Western , Cell Adhesion , Chondrocytes/metabolism , Collagen/genetics , Microscopy, Electron, Scanning , Phenotype , Polyethylene Glycols/chemistry , Polymerase Chain Reaction , RNA, Messenger/genetics
19.
Biomaterials ; 27(7): 1043-53, 2006 Mar.
Article En | MEDLINE | ID: mdl-16125219

Optimizing re-differentiation of clinically relevant cell sources on biomaterial substrates in serum containing (S+) and serum-free (SF) media is a key consideration in scaffold-based articular cartilage repair strategies. We investigated whether the adhesion and post-expansion re-differentiation of human chondrocytes could be regulated by controlled changes in substrate surface chemistry and composition in S+ and SF media following gas plasma (GP) treatment. Expanded human nasal chondrocytes were plated on gas plasma treated (GP+) or untreated (GP-) poly(ethylene glycol)-terephthalate-poly(butylene terephthalate) (PEGT/PBT) block co-polymer films with two compositions (low or high PEG content). Total cellularity, cell morphology and immunofluorescent staining of vitronectin (VN) and fibronectin (FN) integrin receptors were evaluated, while post-expansion chondrogenic phenotype was assessed by collagen types I and II mRNA expression. We observed a direct relationship between cellularity, cell morphology and re-differentiation potential. Substrates supporting high cell adhesion and a spread morphology (i.e. GP+ and low PEG content films), resulted in a significantly greater number of cells expressing alpha5beta1 FN to alpha(V)beta3 VN integrin receptors, concomitant with reduced collagen type II/ImRNA gene expression. Substrates supporting low cell adhesion and a spherical morphology (GP- and high PEG content films) promoted chondrocyte re-differentiation indicated by high collagen type II/I gene expression and a low percentage of alpha5beta1 FN integrin expressing cells. This study demonstrates that cell-substrate interactions via alpha5beta1 FN integrin mediated receptors negatively impacts expanded human nasal chondrocyte re-differentiation capacity. GP treatment promotes cell adhesion in S+ media but reverses the ability of low PEG content PEGT/PBT substrates to maintain chondrocyte phenotype. We suggest alternative cell immobilization techniques to GP are necessary for clinical application in articular cartilage repair.


Biocompatible Materials/chemistry , Chondrocytes/cytology , Chondrocytes/physiology , Hyaline Cartilage/cytology , Hyaline Cartilage/physiology , Polyesters/chemistry , Polyethylene Glycols/chemistry , Tissue Engineering/methods , Adult , Cell Culture Techniques/methods , Cell Differentiation , Cell Proliferation , Cell Size , Cells, Cultured , Cells, Immobilized/physiology , Culture Media, Serum-Free , Gases/chemistry , Hot Temperature , Humans , Integrin alpha5beta1/metabolism , Integrin alphaVbeta3/metabolism , Nasal Septum/cytology , Nasal Septum/physiology , Surface Properties
20.
Exp Cell Res ; 310(1): 54-65, 2005 Oct 15.
Article En | MEDLINE | ID: mdl-16112669

Complement C2 receptor inhibitor trispanning (CRIT) is a receptor for the second component of complement and is found in various tissues and hemopoietic cells. On binding to CRIT, C2 cannot be activated to potentially form a variant-C3 convertase as it is rendered non-cleavable by C1s. CRIT thus limits the amount of C3 convertase formed on the cell surface. In this study we have shown, using flow cytometry and immunofluorescence microscopy, that human CRIT undergoes endocytosis from the plasma membrane. The endocytosis, possibly ligand mediated, occurs via clathrin-coated pits as it can be inhibited by prior incubation of cells in hypertonic medium or with chlorpromazine, at 37 degrees C. However, inhibition of endocytosis was not possible after treatment with nystatin, or filipin, inhibitors of caveolae/raft-dependent endocytosis. In the presence of C2 alone, CRIT associates with the adapter protein, beta-arrestin-2, and whether in association with C2 or not, then appears in the perinuclear region, but does not appear to be translocated into the nucleus. Apart from the C3aR and C5aR that internalize the anaphylatoxic peptides, this is the first report of the internalization via the clathrin pathway of a receptor for a complement serum protein.


Carrier Proteins/metabolism , Clathrin-Coated Vesicles/physiology , Complement Inactivator Proteins/physiology , Endocytosis/physiology , Signal Transduction/physiology , Arrestins/metabolism , Carrier Proteins/pharmacology , Clathrin-Coated Vesicles/chemistry , Colchicine/pharmacology , Complement C2/pharmacology , Complement Inactivator Proteins/pharmacology , Filipin/pharmacology , Humans , Jurkat Cells , Ligands , Nystatin/pharmacology , Phosphoproteins/metabolism , Phosphorylation , Tyrosine/chemistry
...