Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 216
Filter
1.
J Mater Chem B ; 12(22): 5513-5524, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38745541

ABSTRACT

BACKGROUND: In the domain of plastic surgery, nasal cartilage regeneration is of significant importance. The extracellular matrix (ECM) from porcine nasal septum cartilage has shown potential for promoting human cartilage regeneration. Nonetheless, the specific biological inductive factors and their pathways in cartilage tissue engineering remain undefined. METHODS: The decellularized matrix derived from porcine nasal septum cartilage (PN-DCM) was prepared using a grinding method. Human umbilical cord mesenchymal stem cells (HuMSCs) were cultured on these PN-DCM scaffolds for 4 weeks without exogenous growth factors to evaluate their chondroinductive potential. Subsequently, proteomic analysis was employed to identify potential biological inductive factors within the PN-DCM scaffolds. RESULTS: Compared to the TGF-ß3-cultured pellet model serving as a positive control, the PN-DCM scaffolds promoted significant deposition of a Safranin-O positive matrix and Type II collagen by HuMSCs. Gene expression profiling revealed upregulation of ACAN, COL2A1, and SOX9. Proteomic analysis identified potential chondroinductive factors in the PN-DCM scaffolds, including CYTL1, CTGF, MGP, ITGB1, BMP7, and GDF5, which influence HuMSC differentiation. CONCLUSION: Our findings have demonstrated that the PN-DCM scaffolds promoted HuMSC differentiation towards a nasal chondrocyte phenotype without the supplementation of exogenous growth factors. This outcome is associated with the chondroinductive factors present within the PN-DCM scaffolds.


Subject(s)
Cell Differentiation , Chondrogenesis , Mesenchymal Stem Cells , Nasal Septum , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Nasal Septum/cytology , Nasal Septum/chemistry , Animals , Swine , Cells, Cultured , Tissue Scaffolds/chemistry , Decellularized Extracellular Matrix/chemistry , Decellularized Extracellular Matrix/pharmacology , Extracellular Matrix/metabolism , Extracellular Matrix/chemistry , Tissue Engineering , Umbilical Cord/cytology
2.
FASEB J ; 35(3): e21191, 2021 03.
Article in English | MEDLINE | ID: mdl-33595884

ABSTRACT

Skin cancer patients often have tumorigenic lesions on their noses. Surgical resection of the lesions often results in nasal cartilage removal. Cartilage grafts taken from other anatomical sites are used for the surgical reconstruction of the nasal cartilage, but donor-site morbidity is a common problem. Autologous tissue-engineered nasal cartilage grafts can mitigate the problem, but commercially available scaffolds define the shape and sizes of the engineered grafts during tissue fabrication. Moreover, the engineered grafts suffer from the inhomogeneous distribution of the functional matrix of cartilage. Advances in 3D bioprinting technology offer the opportunity to engineer cartilages with customizable dimensions and anatomically shaped configurations without the inhomogeneous distribution of cartilage matrix. Here, we report the fidelity of Freeform Reversible Embedding of Suspended Hydrogel (FRESH) bioprinting as a strategy to generate customizable and homogenously distributed functional cartilage matrix engineered nasal cartilage. Using FRESH and in vitro chondrogenesis, we have fabricated tissue-engineered nasal cartilage from combining bovine type I collagen hydrogel and human nasoseptal chondrocytes. The engineered nasal cartilage constructs displayed molecular, biochemical and histological characteristics akin to native human nasal cartilage.


Subject(s)
Bioprinting/methods , Cartilage, Articular/cytology , Chondrocytes/chemistry , Collagen/chemistry , Hydrogels/chemistry , Nasal Septum/cytology , Tissue Engineering/methods , Adult , Cartilage, Articular/physiology , Chondrogenesis , Humans , Male , Tissue Scaffolds/chemistry
3.
Ann Biomed Eng ; 49(6): 1572-1586, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33409853

ABSTRACT

The treatment of cartilage defect remains a challenging issue in clinical practice. Chitosan-based materials have been recognized as a suitable microenvironment for chondrocyte adhesion, proliferation and differentiation forming articular cartilage. The use of nasal chondrocytes to culture articular cartilage on an appropriate scaffold emerged as a promising novel strategy for cartilage regeneration. Beside excellent properties, chitosan lacks in biological activity, such as RGD-sequences. In this work, we have prepared pure and protein-modified chitosan scaffolds of different deacetylation degree and molecular weight as platforms for the culture of sheep nasal chondrocytes. Fibronectin (FN) was chosen as an adhesive protein for the improvement of chitosan bioactivity. Prepared scaffolds were characterised in terms of microstructure, physical and biodegradation properties, while FN interactions with different chitosans were investigated through adsorption-desorption studies. The results indicated faster enzymatic degradation of chitosan scaffolds with lower deacetylation degree, while better FN interactions with material were achieved on chitosan with higher number of amine groups. Histological and immunohistochemical analysis of in vitro engineered cartilage grafts showed presence of hyaline cartilage produced by nasal chondrocytes.


Subject(s)
Cartilage, Articular , Chitosan , Chondrocytes/physiology , Tissue Scaffolds , Adsorption , Aggrecans/metabolism , Animals , Cartilage, Articular/metabolism , Cell Adhesion , Cell Differentiation , Cell Proliferation , Cells, Cultured , Collagen Type II/metabolism , Fibronectins/chemistry , Nasal Septum/cytology , Phenazines/metabolism , Sheep , Tissue Engineering/methods
4.
Tissue Eng Regen Med ; 17(1): 81-90, 2020 02.
Article in English | MEDLINE | ID: mdl-31983036

ABSTRACT

BACKGROUND: Cell-based therapies have been studied for articular cartilage regeneration. Articular cartilage defects have little treatments because articular cartilage was limited regenerative capacity. Damaged articular cartilage is difficult to obtain a successful therapeutic effect. In additionally these articular cartilage defects often cause osteoarthritis. Chondrocyte implantation is a widely available therapy used for regeneration of articular cartilage because this tissue has poor repair capacity after injury. Human nasal septum-drived chondrocytes (hNCs) from the septum show greater proliferation ability and chondrogenic capacity than human articular chondrocytes (hACs), even across different donors with different ages. Moreover, the chondrogenic properties of hNCs can be maintained after extensive culture expansion. METHODS: In this study, 2 dimensional (2D) monolayer cultured hNCs (hNCs-2D) and 3 dimensional (3D) spheroids cultured hNCs (hNCs-3D) were examined for chondrogenic capacity in vitro by PCR and immunofluorescence staining for chondrogenic marker, cell survival during cultured and for cartilage regeneration ability in vivo in a rat osteochondral defect model. RESULTS: hNCs-3D showed higher viability and more uniform morphology than 3D spheroids cultured hACs (hACs-3D) in culture. hNCs-3D also showed greater expression levels of the chondrocyte-specific marker Type II collagen (COL2A1) and sex-determining region Y (SRY)-box 9 (SOX9) than hNCs-2D. hNCs-3D also expressed chondrogenic markers in collagen. Specially, in the osteochondral defect model, implantation of hNCs-3D led to greater chondrogenic repair of focal cartilage defects in rats than implantation of hNCs-2D. CONCLUSION: These data suggest that hNCs-3D are valuable therapeutic agents for repair and regeneration of cartilage defects.


Subject(s)
Cartilage, Articular/cytology , Chondrocytes/cytology , Nasal Septum/cytology , Regeneration , Animals , Cell Proliferation , Cell Survival , Collagen Type II/metabolism , Gene Expression , Humans , Male , Models, Animal , Rats , Rats, Sprague-Dawley , Tissue Engineering
5.
Tissue Eng Regen Med ; 16(6): 605-614, 2019 12.
Article in English | MEDLINE | ID: mdl-31824823

ABSTRACT

Background: In this study, we manufactured a complex of human nasal septal cartilage (hNC) with polycaprolactone (PCL) for transplantation into cartilaginous skeletal defects and evaluated their characteristics. Methods: Nasal septum tissue was obtained from five patients aged ≥ 20 years who were undergoing septoplasty. hNCs were isolated and subcultured for three passages in vitro. To formulate the cell-PCL complex, we used type I collagen as an adhesive between chondrocyte and PCL. Immunofluorescence staining, cell viability and growth in the hNC-PCL complex, and mycoplasma contamination were assessed. Results: hNCs in PCL showed viability ≥ 70% and remained at these levels for 9 h of incubation at 4 °C. Immunostaining of the hNC-PCL complex also showed high expression levels of chondrocyte-specific protein, COL2A1, SOX9, and aggrecan during 24 h of clinically applicable conditions. Conclusion: The hNC-PCL complex may be a valuable therapeutic agent for implantation into injured cartilage tissue, and can be used clinically to repair cartilaginous skeletal defects. From a clinical perspective, it is important to set the short duration of the implantation process to achieve effective functional implantation.


Subject(s)
Cartilage, Articular/physiology , Polyesters/chemistry , Tissue Scaffolds/chemistry , Cell Survival , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Collagen Type II/genetics , Collagen Type II/metabolism , Humans , Nasal Septum/cytology , Printing, Three-Dimensional , Regeneration , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Tissue Engineering , Young Adult
6.
Sci Rep ; 9(1): 12464, 2019 08 28.
Article in English | MEDLINE | ID: mdl-31462660

ABSTRACT

Nasoseptal cartilage has been assumed to be isotropic, unlike the well-defined zonal organization of articular cartilage attributed to postnatal biomechanical loading. We know from clinical experience that malrotation of surgical nasoseptal cartilage grafts can lead to increased graft absorption. Other studies have also suggested directionally dependent compressive stiffness suggesting anisotropy, but morphological investigations are lacking. This study characterizes immature and mature native bovine nasoseptal cartilage using a combination of immunohistochemistry, biomechanical testing and structural imaging. Our findings indicate that there is extensive postnatal synthesis and reorganization of the extracellular matrix in bovine nasoseptal cartilage, independent of joint loading forces responsible for articular cartilage anisotropy. Immature nasoseptal cartilage is more cellular and homogenous compared to the zonal organization of cells and extracellular matrix of mature cartilage. Mature samples also exhibited greater glycosaminoglycan content and type II collagen fibre alignment compared to immature cartilage and this correlates with greater compressive stiffness. Engineered neocartilage often consists of immature, isotropic, homogenous tissue that is unable to meet the functional and mechanical demands when implanted into the native environment. This study demonstrates the importance of anisotropy on biomechanical tissue strength to guide future cartilage tissue engineering strategies for surgical reconstruction.


Subject(s)
Cartilage/cytology , Cartilage/metabolism , Collagen Type II/metabolism , Compressive Strength , Nasal Septum/cytology , Nasal Septum/metabolism , Animals , Anisotropy , Cartilage/surgery , Cattle , Nasal Septum/surgery
7.
Appl Biochem Biotechnol ; 188(4): 942-951, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30740625

ABSTRACT

The aim of this study was to investigate the effect of medium harvested from septal cartilage cells on chondrogenic differentiation of adipose stem cells (hASCs) and to compare/contrast its properties to those of a commonly used standard medium formulation in terms of induction and maintenance of chondrogenic hASCs. Differentiation was carried out under three different conditions: septal cartilage medium-SCM, chondrogenic differentiation medium-CM, and 50:50 mixture of CM/SCM. Mesenchymal stem cells (MSCs) markers were determined by flow cytometry. The cytotoxic and apoptotic effects were determined by MTS and Annexin V assay, respectively. The differentiation status of the cells was confirmed by Alcian blue staining, and quantitative real-time flow cytometry showed that hASCs were positive for MSCs, negative for hematopoietic stem cells and endothelial cell surface markers. According to MTS analysis, the first condition was not toxic at any concentration tested. Annexin V assay revealed that the application of different concentrations of SCM did not result in any cell death. The Alcian blue and gene expression analyses showed that the cells in the SCM group underwent the highest cartilage cell formation. The observed increase in chondrogenesis may offer better treatment options for the cartilage defects seen in nasal septum perforation.


Subject(s)
Adipocytes/cytology , Cartilage/cytology , Cell Differentiation/physiology , Chondrocytes/cytology , Chondrogenesis/physiology , Nasal Cartilages/cytology , Stem Cells/cytology , Cell Differentiation/genetics , Cells, Cultured , Chondrogenesis/genetics , Flow Cytometry , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Nasal Septum/cytology , Real-Time Polymerase Chain Reaction
8.
Biofabrication ; 11(4): 045006, 2019 07 08.
Article in English | MEDLINE | ID: mdl-30743252

ABSTRACT

BACKGROUND: One of the main challenges for extrusion 3D bioprinting is the identification of non-synthetic bioinks with suitable rheological properties and biocompatibility. Our aim was to optimize and compare the printability of crystal, fibril and blend formulations of novel pulp derived nanocellulose bioinks and assess biocompatibility with human nasoseptal chondrocytes. METHODS: The printability of crystalline, fibrillated and blend formulations of nanocellulose was determined by assessing resolution (grid-line assay), post-printing shape fidelity and rheology (elasticity, viscosity and shear thinning characteristics) and compared these to pure alginate bioinks. The optimized nanocellulose-alginate bioink was bioprinted with human nasoseptal chondrocytes to determine cytotoxicity, metabolic activity and bioprinted construct topography. RESULTS: All nanocellulose-alginate bioink combinations demonstrated a high degree of shear thinning with reversible stress softening behavior which contributed to post-printing shape fidelity. The unique blend of crystal and fibril nanocellulose bioink exhibited nano- as well as micro-roughness for cellular survival and differentiation, as well as maintaining the most stable construct volume in culture. Human nasoseptal chondrocytes demonstrated high metabolic activity post printing and adopted a rounded chondrogenic phenotype after prolonged culture. CONCLUSIONS: This study highlights the favorable rheological, swelling and biocompatibility properties of nanocellulose-alginate bioinks for extrusion-based bioprinting.


Subject(s)
Alginates/chemistry , Bioprinting , Cellulose/chemistry , Ink , Nanoparticles/chemistry , Printing, Three-Dimensional , Wood/chemistry , Biomass , Cell Survival , Cellulose/ultrastructure , Chondrocytes/cytology , Chondrocytes/metabolism , Cross-Linking Reagents/chemistry , Humans , L-Lactate Dehydrogenase/metabolism , Nanoparticles/ultrastructure , Nasal Septum/cytology , Rheology , Stress, Mechanical , Viscosity
9.
Stem Cell Res Ther ; 9(1): 94, 2018 04 07.
Article in English | MEDLINE | ID: mdl-29625584

ABSTRACT

BACKGROUND: Upon orthognathic mandibular advancement surgery the adjacent soft tissues can displace the distal bone segment and increase the load on the temporomandibular joint causing loss of its integrity. Remodeling of the condyle and temporal fossa with destruction of condylar cartilage and subchondral bone leads to postsurgical condylar resorption, with arthralgia and functional limitations. Patients with severe lesions are refractory to conservative treatments, leading to more invasive therapies that range from simple arthrocentesis to open surgery and prosthesis. Although aggressive and with a high risk for the patient, surgical invasive treatments are not always efficient in managing the degenerative lesions. METHODS: We propose a regenerative medicine approach using in-vitro expanded autologous cells from nasal septum applied to the first proof-of-concept patient. After the required quality controls, the cells were injected into each joint by arthrocentesis. Results were monitored by functional assays and image analysis using computed tomography. RESULTS: The cell injection fully reverted the condylar resorption, leading to functional and structural regeneration after 6 months. Computed tomography images showed new cortical bone formation filling the former cavity space, and a partial recovery of condylar and temporal bones. The superposition of the condyle models showed the regeneration of the bone defect, reconstructing the condyle original form. CONCLUSIONS: We propose a new treatment of condylar resorption subsequent to orthognathic surgery, presently treated only by alloplastic total joint replacement. We propose an intra-articular injection of autologous in-vitro expanded cells from the nasal septum. The proof-of-concept treatment of a selected patient that had no alternative therapeutic proposal has given promising results, reaching full regeneration of both the condylar cartilage and bone at 6 months after the therapy, which was fully maintained after 1 year. This first case is being followed by inclusion of new patients with a similar pathological profile to complete an ongoing stage I/II study. TRIAL REGISTRATION: This clinical trial is approved by the National Commission of Ethics in Medical Research (CONEP), Brazil, CAAE 12484813.0.0000.5245, and retrospectively registered in the Brazilian National Clinical Trials Registry and in the USA Clinical Trials Registry under the Universal Trial Number (UTN) U1111-1194-6997 .


Subject(s)
Bone Regeneration , Bone Resorption/surgery , Cell Transplantation/methods , Chondrocytes/transplantation , Orthognathic Surgery/methods , Temporomandibular Joint/surgery , Adult , Bone Resorption/pathology , Cells, Cultured , Humans , Male , Nasal Septum/cytology , Temporomandibular Joint/physiology , Transplantation, Autologous
10.
Sci Rep ; 8(1): 436, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29323189

ABSTRACT

Exposure to lipopolysaccharides (LPS) causes extensive neutrophilic inflammation in the airways followed by mucous cell hyperplasia (MCH) that is sustained by the anti-apoptotic protein, Bcl-2. To identify inflammatory factor(s) that are responsible for Bcl-2 expression, we established an organ culture system consisting of airway epithelial tissue from the rat nasal midseptum. The highest Muc5AC and Bcl-2 expression was observed when organ cultures were treated with brochoalveolar lavage (BAL) fluid harvested from rats 10 h post LPS instillation. Further, because BAL harvested from rats depleted of polymorphonuclear cells compared to controls showed increased Bcl-2 expression, analyses of cytokine levels in lavages identified IL-13 as an inducer of Bcl-2 expression. Ectopic IL-13 treatment of differentiated airway epithelial cells increased Bcl-2 and MUC5AC expression in the basal and apical regions of the cells, respectively. When Bcl-2 was blocked using shRNA or a small molecule inhibitor, ABT-263, mucous cell numbers were reduced due to increased apoptosis that disrupted the interaction of Bcl-2 with the pro-apoptotic protein, Bik. Furthermore, intranasal instillation of ABT-263 reduced the LPS-induced MCH in bik +/+ but not bik -/- mice, suggesting that Bik mediated apoptosis in hyperplastic mucous cells. Therefore, blocking Bcl-2 function could be useful in reducing IL-13 induced mucous hypersecretion.


Subject(s)
Inflammation/metabolism , Interleukin-13/metabolism , Lipopolysaccharides/adverse effects , Nasal Septum/cytology , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Apoptosis , Bronchoalveolar Lavage Fluid/immunology , Hyperplasia , Male , Mucin 5AC/metabolism , Nasal Septum/metabolism , Nasal Septum/pathology , Organ Culture Techniques , Rats
11.
Cell Tissue Bank ; 19(1): 35-46, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28815373

ABSTRACT

Cell based tissue engineering serves as a promising strategy for articular cartilage repair, which remains a challenge both for researchers and clinicians. The aim of this research was to assess the potential of autologous nasal chondrocytes (NCs) combined with alginate hydrogel as injectable constructs for rabbit articular cartilage repair. Autologous nasal chondrocytes were isolated from rabbit nasal septum, expanded either on monolayer or in 3D alginate hydrogel. In vitro, DNA quantification revealed that NCs can proliferate stable in 3D alginate matrix, but slower than that cultured in monolayer. Further, a higher synthesis rate of glycosaminoglycans (GAGs) was detected by GAG measurement in 3D alginate culture. Gene expression analysis at different time point (day 1, 7, 14) showed that 3D culture of NCs in alginate up-regulated chondrogenic markers (Col2A1, ACAN SOX9), meanwhile down-regulated dedifferentiation related gene (Col1A1). In vivo, autologous nasal chondrocytes combined with alginate hydrogel were used for repairing rabbit knee osteochondral defect (Alg + NC group). Histological staining indicated that Alg + NC group obtained superior and more hyaline-like repaired tissue both at 3 and 6 months after surgery. Mechanical analysis showed that the repaired tissue in the Alg + NC group possessed similar mechanical properties to the native cartilage. In conclusion, nasal chondrocytes appeared to be a very promising seed cell source for cartilage tissue engineering, and alginate hydrogel can serve as suitable delivery system.


Subject(s)
Cartilage, Articular/physiology , Chondrocytes/transplantation , Hydrogels/chemistry , Nasal Septum/cytology , Regeneration , Tissue Scaffolds/chemistry , Alginates/administration & dosage , Animals , Cartilage, Articular/cytology , Cell Separation , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrogenesis , Glucuronic Acid/administration & dosage , Hexuronic Acids/administration & dosage , Hydrogels/administration & dosage , Injections , Rabbits , Tissue Engineering
12.
J Tissue Eng Regen Med ; 12(4): 1123-1132, 2018 04.
Article in English | MEDLINE | ID: mdl-29139602

ABSTRACT

Functional reconstruction of large cartilage defects in subcutaneous sites remains clinically challenging because of limited donor cartilage. Tissue engineering is a promising and widely accepted strategy for cartilage regeneration. To date, however, this strategy has not achieved a significant breakthrough in clinical translation owing to a lack of detailed preclinical data on cell yield and functionality of clinically applicable chondrocytes. To address this issue, the current study investigated the initial cell yield, proliferative potential, chondrogenic capacity, and regenerated cartilage type of human chondrocytes derived from auricular, nasoseptal, and costal cartilage using a scaffold-free cartilage regeneration model (cartilage sheet). Chondrocytes from all sources exhibited high sensitivity to basic fibroblast growth factor within 8 passages. Nasoseptal chondrocytes presented the strongest proliferation rate, whereas auricular chondrocytes obtained the highest total cell amount using comparable cartilage sample weights. Importantly, all chondrocytes at fifth passage showed strong chondrogenic capacity both in vitro and in the subcutaneous environment of nude mice. Although some significant differences in histological structure, cartilage matrix content and cartilage type specific proteins were observed between the in vitro engineered cartilage and original tissue; the in vivo regenerated cartilage showed mature cartilage features with high similarity to their original native tissue, except for minor matrix changes influenced by the in vivo environment. The current study provides detailed preclinical data for choice of chondrocyte source and thus promotes the clinical translation of cartilage regeneration approach.


Subject(s)
Cell Separation , Chondrocytes , Chondrogenesis , Costal Cartilage/cytology , Ear Cartilage/cytology , Nasal Septum/cytology , Animals , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrocytes/transplantation , Costal Cartilage/metabolism , Ear Cartilage/metabolism , Humans , Mice, Nude , Nasal Septum/metabolism
13.
Biomed Res Int ; 2017: 2395138, 2017.
Article in English | MEDLINE | ID: mdl-28337445

ABSTRACT

Laboratory based processing and expansion to yield adequate cell numbers had been the standard in Autologous Disc Chondrocyte Transplantation (ADCT), Allogeneic Juvenile Chondrocyte Implantation (NuQu®), and Matrix-Induced Autologous Chondrocyte Implantation (MACI). Optimizing cell isolation is a key challenge in terms of obtaining adequate cell numbers while maintaining a vibrant cell population capable of subsequent proliferation and matrix elaboration. However, typical cell yields from a cartilage digest are highly variable between donors and based on user competency. The overall objective of this study was to optimize chondrocyte isolation from cartilaginous nasal tissue through modulation of enzyme concentration exposure (750 and 3000 U/ml) and incubation time (1 and 12 h), combined with physical agitation cycles, and to assess subsequent cell viability and matrix forming capacity. Overall, increasing enzyme exposure time was found to be more detrimental than collagenase concentration for subsequent viability, proliferation, and matrix forming capacity (sGAG and collagen) of these cells resulting in nonuniform cartilaginous matrix deposition. Taken together, consolidating a 3000 U/ml collagenase digest of 1 h at a ratio of 10 ml/g of cartilage tissue with physical agitation cycles can improve efficiency of chondrocyte isolation, yielding robust, more uniform matrix formation.


Subject(s)
Cartilage/cytology , Cell Separation , Chondrocytes/cytology , Tissue Engineering , Animals , Cartilage/growth & development , Cartilage/transplantation , Cattle , Cell Proliferation/drug effects , Chondrocytes/transplantation , Collagenases/administration & dosage , Humans , Nasal Septum/cytology , Nasal Septum/transplantation , Transplantation, Autologous/methods , Transplantation, Homologous/methods
14.
Laryngoscope ; 127(3): E91-E99, 2017 03.
Article in English | MEDLINE | ID: mdl-27861930

ABSTRACT

OBJECTIVE: Cartilage tissue engineering is a promising approach to provide suitable materials for nasal reconstruction; however, it typically requires large numbers of cells. We have previously shown that a small number of chondrocytes cultivated within a continuous flow bioreactor can elicit substantial tissue growth, but translation to human chondrocytes is not trivial. Here, we aimed to demonstrate the application of the bioreactor to generate large-sized tissues from a small population of primary human nasoseptal chondrocytes. STUDY DESIGN: Experimental study. METHODS: Chondrocytes were cultured in the bioreactor using different medium compositions, with varying amounts of serum and with or without growth factors. Resulting engineered tissues were analyzed for physical properties, biochemical composition, tissue microstructure, and protein localization. RESULTS: Bioreactor-cultivated constructs grown with serum and growth factors (basic fibroblast growth factor and transforming growth factor beta 2) had greater thickness, as well as DNA and glycosaminoglycan (GAG) contents, compared to low serum and no growth factor controls. These constructs also showed the most intense proteoglycan and collagen II staining. CONCLUSION: The combination of bioreactor conditions, serum, and growth factors allowed the generation of large, thick scaffold-free human cartilaginous tissues that resembled the native nasoseptal cartilage. There also may be implications for patient selection in future clinical applications of these engineered tissues because their GAG content decreased with donor age. LEVEL OF EVIDENCE: NA. Laryngoscope, 127:E91-E99, 2017.


Subject(s)
Chondrocytes/cytology , Tensile Strength , Tissue Engineering/methods , Biomechanical Phenomena , Cell Culture Techniques , Cells, Cultured , Chondrocytes/pathology , Fibroblast Growth Factor 2/administration & dosage , Humans , Immunohistochemistry , Nasal Septum/cytology , Nasal Surgical Procedures/methods , Receptors, Transforming Growth Factor beta/administration & dosage , Plastic Surgery Procedures/methods , Tissue Scaffolds , Tissue and Organ Harvesting
15.
Lancet ; 388(10055): 1985-1994, 2016 Oct 22.
Article in English | MEDLINE | ID: mdl-27789021

ABSTRACT

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.


Subject(s)
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
16.
Tissue Eng Part A ; 22(21-22): 1286-1295, 2016 11.
Article in English | MEDLINE | ID: mdl-27633049

ABSTRACT

Nasal chondrocytes (NC) were previously demonstrated to remain viable and to participate in the repair of articular cartilage defects in goats. Here, we investigated critical features of tissue-engineered grafts generated by NC in this large animal model, namely cell retention at the implantation site, architecture and integration with adjacent tissues, and effects on subchondral bone changes. In this study, isolated autologous goat NC (gNC) and goat articular chondrocytes (gAC, as control) were expanded, green fluorescent protein-labelled and seeded on a type I/III collagen membrane. After chondrogenic differentiation, tissue-engineered grafts were implanted into chondral defects (6 mm in diameter) in the stifle joint for 3 or 6 months. At the time of explantation, surrounding tissues showed no or very low (only in the infrapatellar fat pad <0.32%) migration of the grafted cells. In repair tissue, gNC formed typical structures of articular cartilage, such as flattened cells at the surface and column-like clusters in the middle layers. Semi-quantitative histological evaluation revealed efficient integration of the grafted tissues with the adjacent native cartilage and underlying subchondral bone. A significantly increased subchondral bone area, as a sign for the onset of osteoarthritis, was observed following treatment of cartilage defects with gAC-, but not with gNC-grafts. Our results reinforce the use of NC-based engineered tissue for articular cartilage repair and preliminarily indicate their potential for the treatment of early osteoarthritic defects.


Subject(s)
Cartilage, Articular , Chondrocytes/metabolism , Nasal Septum , Regeneration , Tissue Engineering , Animals , Cartilage, Articular/injuries , Cartilage, Articular/physiology , Chondrocytes/cytology , Chondrocytes/transplantation , Female , Goats , Nasal Septum/cytology , Nasal Septum/metabolism
17.
Laryngoscope ; 126(10): 2226-31, 2016 10.
Article in English | MEDLINE | ID: mdl-27232879

ABSTRACT

OBJECTIVES/HYPOTHESIS: Reconstruction of cartilage defects in the head and neck can require harvesting of autologous cartilage grafts, which can be associated with donor site morbidity. To overcome this limitation, tissue-engineering approaches may be used to generate cartilage grafts. The objective of this study was to decellularize and characterize human nasoseptal cartilage with the aim of generating a biological scaffold for cartilage tissue engineering. STUDY DESIGN: Laboratory study using nasoseptal cartilage. METHODS: Remnant human nasoseptal cartilage specimens were collected and subjected to a novel decellularization treatment. The decellularization process involved several cycles of enzymatic detergent treatments. For characterization, decellularized and fresh (control) specimens underwent histological, biochemical, and mechanical analyses. Scanning electron microscopy and biocompatibility assay were also performed. RESULTS: The decellularization process had minimal effect on glycosaminoglycan content of the cartilage extracellular matrix. Deoxyribonucleic acid (DNA) analysis revealed the near-complete removal of genomic DNA from decellularized tissues. The effectiveness of the decellularization process was also confirmed on histological and scanning electron microscopic analyses. Mechanical testing results showed that the structural integrity of the decellularized tissue was maintained, and biocompatibility was confirmed. CONCLUSION: Overall, the current decellularization treatment resulted in significant reduction of genetic/cellular material with preservation of the underlying extracellular matrix structure. This decellularized material may serve as a potential scaffold for cartilage tissue engineering. LEVEL OF EVIDENCE: N/A. Laryngoscope, 126:2226-2231, 2016.


Subject(s)
Cartilage/cytology , Extracellular Matrix/chemistry , Nasal Septum/cytology , Tissue Engineering/methods , Cartilage/transplantation , Cell-Free System , DNA/analysis , Glycosaminoglycans/analysis , Humans , Tissue Scaffolds
18.
J Allergy Clin Immunol ; 138(3): 825-838, 2016 09.
Article in English | MEDLINE | ID: mdl-26971690

ABSTRACT

BACKGROUND: IgG4-related disease (IgG4-RD) is a systemic condition of unknown cause characterized by highly fibrotic lesions with dense lymphoplasmacytic infiltrates. CD4(+) T cells constitute the major inflammatory cell population in IgG4-RD lesions. OBJECTIVE: We used an unbiased approach to characterize CD4(+) T-cell subsets in patients with IgG4-RD based on their clonal expansion and ability to infiltrate affected tissue sites. METHODS: We used flow cytometry to identify CD4(+) effector/memory T cells in a cohort of 101 patients with IgG4-RD. These expanded cells were characterized by means of gene expression analysis and flow cytometry. Next-generation sequencing of the T-cell receptor ß chain gene was performed on CD4(+)SLAMF7(+) cytotoxic T lymphocytes (CTLs) and CD4(+)GATA3(+) TH2 cells in a subset of patients to identify their clonality. Tissue infiltration by specific T cells was examined by using quantitative multicolor imaging. RESULTS: CD4(+) effector/memory T cells with a cytolytic phenotype were expanded in patients with IgG4-RD. Next-generation sequencing revealed prominent clonal expansions of these CD4(+) CTLs but not CD4(+)GATA3(+) memory TH2 cells in patients with IgG4-RD. The dominant T cells infiltrating a range of inflamed IgG4-RD tissue sites were clonally expanded CD4(+) CTLs that expressed SLAMF7, granzyme A, IL-1ß, and TGF-ß1. Clinical remission induced by rituximab-mediated B-cell depletion was associated with a reduction in numbers of disease-associated CD4(+) CTLs. CONCLUSIONS: IgG4-RD is prominently linked to clonally expanded IL-1ß- and TGF-ß1-secreting CD4(+) CTLs in both peripheral blood and inflammatory tissue lesions. These active, terminally differentiated, cytokine-secreting effector CD4(+) T cells are now linked to a human disease characterized by chronic inflammation and fibrosis.


Subject(s)
Immune System Diseases/immunology , Immunoglobulin G/immunology , T-Lymphocytes, Cytotoxic/immunology , Adult , Aged , Cytokines/immunology , Female , Humans , Immune System Diseases/blood , Immunoglobulin G/blood , Kidney/cytology , Lung/cytology , Lymph Nodes/cytology , Lymphocyte Count , Male , Middle Aged , Nasal Septum/cytology , Retroperitoneal Space , Submandibular Gland/cytology
19.
J Anat ; 228(1): 113-24, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26552989

ABSTRACT

A long rostrum has distinct advantages for prey capture in an aquatic or semi-aquatic environment but at the same time poses severe problems concerning stability during biting. We here investigate the role of the septum nasi of brevirostrine crocodilians for load-absorption during mastication. Histologically, both the septum nasi and the septum interorbitale consist of hyaline cartilage and therefore mainly resist compression. However, we identified a strand of tissue extending longitudinally below the septum nasi that is characterized by a high content of collagenous and elastic fibers and could therefore resist tensile stresses. This strand of tissue is connected with the m. pterygoideus anterior. Two-dimensional finite element modeling shows that minimization of bending in the crocodilian skull can only be achieved if tensile stresses are counteracted by a strand of tissue. We propose that the newly identified strand of tissue acts as an active tension chord necessary for stabilizing the long rostrum of crocodilians during biting by transforming the high bending stress of the rostrum into moderate compressive stress.


Subject(s)
Alligators and Crocodiles/anatomy & histology , Cartilage/physiology , Mastication/physiology , Nasal Septum/anatomy & histology , Animals , Bite Force , Chondrocytes/cytology , Compressive Strength/physiology , Finite Element Analysis , Nasal Septum/cytology , Nasal Septum/physiology , Tensile Strength/physiology
20.
Ann Biomed Eng ; 44(5): 1821-31, 2016 May.
Article in English | MEDLINE | ID: mdl-26502171

ABSTRACT

Bovine nasal septum (BNS) is a source of non-load bearing hyaline cartilage. Little information is available on its mechanical and biological properties. The aim of this work was to assess the characteristics of BNS cartilage and investigate its behavior in in vitro mechanobiological experiments. Mechanical tests, biochemical assays, and microscopic assessment were performed for tissue characterization. Compressions tests showed that the tissue is viscoelastic, although values of elastic moduli differ from the ones of other cartilaginous tissues. Water content was 78 ± 1.4%; glycosaminoglycans and collagen contents-measured by spectrophotometric assay and hydroxyproline assay-were 39 ± 5% and 25 ± 2.5% of dry weight, respectively. Goldner's Trichrome staining and transmission electron microscopy proved isotropic cells distribution and results of earlier cell division. Furthermore, gene expression was measured after uniaxial compression, showing variations depending on compression time as well as trends depending on equilibration time. In conclusion, BNS has been characterized at several levels, revealing that bovine nasal tissue is regionally homogeneous. Results suggest that, under certain conditions, BNS could be used to perform in vitro cartilage loading experiments.


Subject(s)
Cartilage , Compressive Strength/physiology , Elastic Modulus , Nasal Septum , Animals , Cartilage/cytology , Cartilage/metabolism , Cattle , Nasal Septum/cytology , Nasal Septum/metabolism , Weight-Bearing/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...