Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Orthop Translat ; 45: 140-154, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38559899

ABSTRACT

Background: Cartilage tissue engineering faces challenges related to the use of scaffolds and limited seed cells. This study aims to propose a cost-effective and straightforward approach using costal chondrocytes (CCs) as an alternative cell source to overcome these challenges, eliminating the need for special culture equipment or scaffolds. Methods: CCs were cultured at a high cell density with and without ascorbic acid treatment, serving as the experimental and control groups, respectively. Viability and tissue-engineered constructs (TEC) formation were evaluated until day 14. Slices of TEC samples were used for histological staining to evaluate the secretion of glycosaminoglycans and different types of collagen proteins within the extracellular matrix. mRNA sequencing and qPCR were performed to examine gene expression related to cartilage matrix secretion in the chondrocytes. In vivo experiments were conducted by implanting TECs from different groups into the defect site, followed by sample collection after 12 weeks for histological staining and scoring to evaluate the extent of cartilage regeneration. Hematoxylin-eosin (HE), Safranin-O-Fast Green, and Masson's trichrome stainings were used to examine the content of cartilage-related matrix components in the in vivo repair tissue. Immunohistochemical staining for type I and type II collagen, as well as aggrecan, was performed to assess the presence and distribution of these specific markers. Additionally, immunohistochemical staining for type X collagen was used to observe any hypertrophic changes in the repaired tissue. Results: Viability of the chondrocytes remained high throughout the culture period, and the TECs displayed an enriched extracellular matrix suitable for surgical procedures. In vitro study revealed glycosaminoglycan and type II collagen production in both groups of TEC, while the TEC matrix treated with ascorbic acid displayed greater abundance. The results of mRNA sequencing and qPCR showed that genes related to cartilage matrix secretion such as Sox9, Col2, and Acan were upregulated by ascorbic acid in costal chondrocytes. Although the addition of Asc-2P led to an increase in COL10 expression according to qPCR and RNA-seq results, the immunofluorescence staining results of the two groups of TECs exhibited similar distribution and fluorescence intensity. In vivo experiments showed that both groups of TEC could adhere to the defect sites and kept hyaline cartilage morphology until 12 weeks. TEC treated with ascorbic acid showed superior cartilage regeneration as evidenced by significantly higher ICRS and O'Driscoll scores and stronger Safranin-O and collagen staining mimicking native cartilage when compared to other groups. In addition, the immunohistochemical staining results of Collgan X indicated that, after 12 weeks, the ascorbic acid-treated TEC did not exhibit further hypertrophy upon transplantation into the defect site, but maintained an expression profile similar to untreated TECs, while slightly higher than the sham-operated group. Conclusion: These results suggest that CC-derived scaffold-free TEC presents a promising method for articular cartilage regeneration. Ascorbic acid treatment enhances outcomes by promoting cartilage matrix production. This study provides valuable insights and potential advancements in the field of cartilage tissue engineering. The translational potential of this article: Cartilage tissue engineering is an area of research with immense clinical potential. The approach presented in this article offers a cost-effective and straightforward solution, which can minimize the complexity of cell culture and scaffold fabrication. This simplification could offer several translational advantages, such as ease of use, rapid scalability, lower costs, and the potential for patient-specific clinical translation. The use of costal chondrocytes, which are easily obtainable, and the scaffold-free approach, which does not require specialized equipment or membranes, could be particularly advantageous in clinical settings, allowing for in situ regeneration of cartilage.

2.
Ann Biomed Eng ; 52(4): 920-933, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38190025

ABSTRACT

This study aimed to compare the histological, biochemical, and mechanical characteristics of hyaline cartilage in different regions and evaluate the potential of chondrocytes extracted from each region as donor sources for articular cartilage repair. The cartilage tissues of the femoral head and knee joint, ribs, nasal septum, thyroid, and xiphoid process of adult Bama pigs were isolated for histological, biochemical, and mechanical evaluation and analysis. The corresponding chondrocytes were isolated and evaluated for proliferation and redifferentiation capacity, using biochemical and histological analysis and RT-PCR experiments. Compared with articular cartilage, non-articular hyaline cartilage matrix stained more intensely in Safranin-O staining. Glycosaminoglycan and total collagen content were similar among all groups, while the highest content was measured in nasal septal cartilage. Regarding biomechanics, non-articular cartilage is similar to articular cartilage, but the elastic modulus and hardness are significantly higher in the middle region of costal cartilage. The chondrocytes extracted from different regions had no significant difference in morphology. Hyaline cartilage-like pellets were formed in each group after redifferentiation. The RT-PCR results revealed similar expressions of cartilage-related genes across the groups, albeit with lower expression of Col2 in the xiphoid chondrocytes. Conversely, higher expression of Col10 was observed in the chondrocytes from the rib, thyroid, and xiphoid cartilage. This study provides valuable preclinical data for evaluating heterotopic hyaline cartilage and chondrocytes for articular cartilage regeneration. The findings contribute to the selection of chondrocyte origins and advance the clinical translation of technology for cartilage regeneration.


Subject(s)
Cartilage, Articular , Swine , Animals , Hyaline Cartilage , Chondrocytes , Knee Joint , Biomechanical Phenomena
3.
Cartilage ; : 19476035231209404, 2023 Oct 26.
Article in English | MEDLINE | ID: mdl-37881954

ABSTRACT

OBJECTIVE: Osteochondral defects develop into osteoarthritis without intervention. Costal cartilage can be utilized as an alternative source for repairing osteochondral defect. Our previous clinical study has shown the successful osteochondral repair by costal cartilage graft with integration into host bone bed. In this study, we investigate how cartilaginous graft adapt to osteochondral environment and the mechanism of bone-cartilage interface formation. DESIGN: Costal cartilage grafting was performed in C57BL/6J mice and full-thickness osteochondral defect was made as control. 3D optical profiles and micro-CT were applied to evaluate the reconstruction of articular cartilage surface and subchondral bone as well as gait analysis to evaluate articular function. Histological staining was performed at 2, 4, and 8 weeks after surgery. Moreover, costal cartilage from transgenic mice with fluorescent markers were transplanted into wild-type mice to observe the in vivo changes of costal chondrocytes. RESULTS: At 8 weeks after surgery, 3D optical profiles and micro-CT showed that in the graft group, the articular surface and subchondral bone were well preserved. Gait analysis and International Cartilage Repair Society (ICRS) score evaluation showed a good recovery of joint function and histological repair in the graft group. Safranin O staining showed the gradual integration of graft and host tissue. Costal cartilage from transgenic mice with fluorescent markers showed that donor-derived costal chondrocytes turned into osteocytes in the subchondral area of host femur. CONCLUSION: Costal cartilage grafting shows both functional and histological repair of osteochondral defect in mice. Graft-derived costal chondrocytes differentiate into osteocytes and contribute to endochondral ossification.

4.
J Nanobiotechnology ; 21(1): 341, 2023 Sep 22.
Article in English | MEDLINE | ID: mdl-37736726

ABSTRACT

Osteoarthritis (OA) is a degenerative joint disease involving cartilage. Exosomes derived from Mesenchymal stem cells (MSCs) therapy improves articular cartilage repair, but subcutaneous fat (SC) stromal cells derived exosomes (MSCsSC-Exos), especially engineering MSCsSC-Exos for drug delivery have been rarely reported in OA therapy. This objective of this study was to clarify the underlying mechanism of MSCsSC-Exos on cartilage repair and therapy of engineering MSCsSC-Exos for drug delivery in OA. MSCsSC-Exos could ameliorate the pathological severity degree of cartilage via miR-199a-3p, a novel molecular highly enriched in MSCsSC-Exos, which could mediate the mTOR-autophagy pathway in OA rat model. Intra-articular injection of antagomiR-199a-3p dramatically attenuated the protective effect of MSCsSC-Exos-mediated on articular cartilage in vivo. Furthermore, to achieve the superior therapeutic effects of MSCsSC-Exos on injured cartilage, engineering exosomes derived from MSCsSC as the chondrocyte-targeting miR-199a-3p delivery vehicles were investigated in vitro and in vivo. The chondrocyte-binding peptide (CAP) binding MSCsSC-Exos could particularly deliver miR-199a-3p into the chondrocytes in vitro and into deep articular tissues in vivo, then exert the excellent protective effect on injured cartilage in DMM-induced OA mice. As it is feasible to obtain human subcutaneous fat from healthy donors by liposuction operation in clinic, meanwhile engineering MSCsSC-Exos to realize targeted delivery of miR-199a-3p into chondrocytes exerted excellent therapeutic effects in OA animal model in vivo. Through combining MSCsSC-Exos therapy and miRNA therapy via an engineering approach, we develop an efficient MSCsSC-Exos-based strategy for OA therapy and promote the application of targeted-MSCsSC-Exos for drug delivery in the future.


Subject(s)
Exosomes , Mesenchymal Stem Cells , MicroRNAs , Osteoarthritis , Humans , Animals , Mice , Rats , MicroRNAs/genetics , Subcutaneous Fat , Osteoarthritis/therapy
5.
Int Immunopharmacol ; 120: 110390, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37262955

ABSTRACT

BACKGROUND: Osteoarthritis (OA) is a highly degenerative joint disease, mainly companying with progressive destruction of articular cartilage. Adipose-derived stromal cells (ADSCs) therapy enhances articular cartilage repair, extracellular matrix (ECM) synthesis and attenuates joints inflammation, but specific mechanisms of therapeutic benefit remain poorly understood. This study aimed to clarify the therapeutic effects and mechanisms of ADSCs on cartilage damage in the keen joint of OA rat model. METHODS: Destabilization of the medial meniscus (DMM) and anterior cruciate ligament transection (ACLT) surgery-induced OA rats were treated with allogeneic ADSCs by intra-articular injections for 6 weeks. The protective effect of ADSCs in vivo was measured using Safranin O and fast green staining, immunofluorescence and western blot analysis. Meanwhile, the miRNA-7-5p (miR-7-5p) expression was assessed by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The mechanism of increased autophagy with ADSCs addition through decreasing miR-7-5p was revealed using oligonucleotides, and adenovirus in rat chondrocytes. The luciferase reporter assay revealed the molecular role of miR-7-5p and autophagy related 4A (ATG4A). The substrate of mTORC1 pathway: (p-)p70S6 and (p-)S6 in OA models with ADSCs addition were detected by western blotting. RESULTS: The ADSCs treatment repaired the articular cartilage and maintained chondrocytes ECM homeostasis through modulating chondrocytes autophagy in the OA model, indicators of the change of autophagic proteins expression and autophagic flux. Meanwhile, the increased autophagy induced by ADSCs treatment was closely related to the decreased expression of host-derived miR-7-5p, a negative modulator of OA progression. Functional genomics (overexpression of genes) in vitro studies demonstrate the inhibition of host-derived miR-7-5p in mediating the benefit of ADSCs administration in OA model. Then ATG4A was defined as a target gene of miR-7-5p, and the negative relation between miR-7-5p and ATG4A was investigated in the OA model treated with ADSCs. Furthermore, miR-7-5p mediated chondrocyte autophagy by targeting ATG4A in the OA model treated with ADSCs was confirmed with the rescue trial of ATG4A/miR-7-5p overexpression on rat chondrocyte. Finally, the mTORC1 signaling pathways mediated by host-derived miR-7-5p with ADSCs treatment were decreased in OA rats. CONCLUSIONS: ADSCs promote the chondrocytes autophagy by decreasing miR-7-5p in articular cartilage by targeting ATG4A and a potential role for ADSCs based therapeutics for preventing of articular cartilage destruction and extracellular matrix (ECM) degradation in OA.


Subject(s)
Cartilage, Articular , MicroRNAs , Osteoarthritis , Rats , Animals , MicroRNAs/metabolism , Chondrocytes , Osteoarthritis/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Cartilage, Articular/metabolism , Autophagy , Apoptosis
6.
J Bone Joint Surg Am ; 104(23): 2108-2116, 2022 12 07.
Article in English | MEDLINE | ID: mdl-36325763

ABSTRACT

BACKGROUND: There is currently no ideal treatment for osteochondral lesions of the femoral head (OLFH) in young patients. METHODS: We performed a 1-year single-arm study and 2 additional years of follow-up of patients with a large (defined as >3 cm 2 ) OLFH treated with insertion of autologous costal cartilage graft (ACCG) to restore femoral head congruity after lesion debridement. Twenty patients ≤40 years old who had substantial hip pain and/or dysfunction after nonoperative treatment were enrolled at a single center. The primary outcome was the change in Harris hip score (HHS) from baseline to 12 months postoperatively. Secondary outcomes included the EuroQol visual analogue scale (EQ VAS), hip joint space width, subchondral integrity on computed tomography scanning, repair tissue status evaluated with the Magnetic Resonance Observation of Cartilage Repair Tissue (MOCART) score, and evaluation of cartilage biochemistry by delayed gadolinium-enhanced magnetic resonance imaging of cartilage (dGEMRIC) and T2 mapping. RESULTS: All 20 enrolled patients (31.02 ± 7.19 years old, 8 female and 12 male) completed the initial study and the 2 years of additional follow-up. The HHS improved from 61.89 ± 6.47 at baseline to 89.23 ± 2.62 at 12 months and 94.79 ± 2.72 at 36 months. The EQ VAS increased by 17.00 ± 8.77 at 12 months and by 21.70 ± 7.99 at 36 months (p < 0.001 for both). Complete integration of the ACCG with the bone was observed by 12 months in all 20 patients. The median MOCART score was 85 (interquartile range [IQR], 75 to 95) at 12 months and 75 (IQR, 65 to 85) at the last follow-up (range, 24 to 38 months). The ACCG demonstrated magnetic resonance properties very similar to hyaline cartilage; the median ratio between the relaxation times of the ACCG and recipient cartilage was 0.95 (IQR, 0.90 to 0.99) at 12 months and 0.97 (IQR, 0.92 to 1.00) at the last follow-up. CONCLUSIONS: ACCG is a feasible method for improving hip function and quality of life for at least 3 years in young patients who were unsatisfied with nonoperative treatment of an OLFH. Promising long-term outcomes may be possible because of the good integration between the recipient femoral head and the implanted ACCG. LEVEL OF EVIDENCE: Therapeutic Level IV . See Instructions for Authors for a complete description of levels of evidence.


Subject(s)
Costal Cartilage , Humans , Female , Male , Adult , Young Adult , Femur Head/diagnostic imaging , Femur Head/surgery , Quality of Life
7.
Stem Cell Res Ther ; 13(1): 386, 2022 07 30.
Article in English | MEDLINE | ID: mdl-35907866

ABSTRACT

BACKGROUND: Seeding cells are key factors in cell-based cartilage tissue regeneration. Monoculture of either chondrocyte or mesenchymal stem cells has several limitations. In recent years, co-culture strategies have provided potential solutions. In this study, directly co-cultured rat costal chondrocytes (CCs) and human Wharton's jelly mesenchymal stem (hWJMSCs) cells were evaluated as a candidate to regenerate articular cartilage. METHODS: Rat CCs are directly co-cultured with hWJMSCs in a pellet model at different ratios (3:1, 1:1, 1:3) for 21 days. The monoculture pellets were used as controls. RT-qPCR, biochemical assays, histological staining and evaluations were performed to analyze the chondrogenic differentiation of each group. The 1:1 ratio co-culture pellet group together with monoculture controls were implanted into the osteochondral defects made on the femoral grooves of the rats for 4, 8, 12 weeks. Then, macroscopic and histological evaluations were performed. RESULTS: Compared to rat CCs pellet group, 3:1 and 1:1 ratio group demonstrated similar extracellular matrix production but less hypertrophy intendency. Immunochemistry staining found the consistent results. RT-PCR analysis indicated that chondrogenesis was promoted in co-cultured rat CCs, while expressions of hypertrophic genes were inhibited. However, hWJMSCs showed only slightly improved in chondrogenesis but not significantly different in hypertrophic expressions. In vivo experiments showed that all the pellets filled the defects but co-culture pellets demonstrated reduced hypertrophy, better surrounding cartilage integration and appropriate subchondral bone remodeling. CONCLUSION: Co-culture of rat CCs and hWJMSCs demonstrated stable chondrogenic phenotype and decreased hypertrophic intendency in both vitro and vivo. These results suggest this co-culture combination as a promising candidate in articular cartilage regeneration.


Subject(s)
Cartilage, Articular , Mesenchymal Stem Cells , Wharton Jelly , Animals , Cartilage, Articular/pathology , Cell Differentiation , Cells, Cultured , Chondrocytes/metabolism , Chondrogenesis/genetics , Coculture Techniques , Humans , Hypertrophy/metabolism , Mesenchymal Stem Cells/metabolism , Rats , Tissue Engineering/methods
8.
Front Pediatr ; 10: 914889, 2022.
Article in English | MEDLINE | ID: mdl-35859948

ABSTRACT

Introduction: Mucopolysaccharidosis Type IVA (MPS IVA) or Morquio A Syndrome, is a rare metabolic disorder caused by compromised galactosamine-6 sulfatase (GALNS) encoded by GALNS gene (NM_000512.5), leading to keratin sulfate (KS), and chondroitin-6-sulfate accumulation in various organs. We present a 17-year-old woman with progressive bilateral hip pain and radiographic evidence of spondyloepiphyseal dysplasia. Methods: Diagnosis of MPS IVA was made based on whole-exome sequencing (WES) of blood samples collected from the patient and family members, high urinary glycosaminoglycan excretion, supportive clinical manifestations, radiographic examinations, including whole-body X-rays, cervical MRI, and pelvic CT. The patient underwent bilateral total hip arthroplasties sequentially, at a 1-month interval. Femoral heads were preserved for the micro-CT (µCT) analysis and the osteochondral histology examination. Results: The patient presented with multiple skeletal deformities, including vertebras and long bone deformities. WES disclosed compound heterozygous variants at exon 11 (c.1156C>T) and exon 12 (c.1288C>G) of the GALNS (NM_000512.5). The µCT analysis revealed significant bone quantity loss and microarchitectural change in both weight-bearing area (WBA) and non-weight-bearing area (NWBA) of the femoral heads, while histological analysis showed structural abnormity of articular cartilage in the WBA of the femoral heads. Conclusion: We have found compound heterozygous variants of GALNS. This is also the first study to report the microarchitectural and histological changes of both subchondral bone and articular cartilage of the femoral head in a patient with MPS IVA.

9.
J Orthop Translat ; 32: 59-68, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34934627

ABSTRACT

BACKGROUND: Costal chondrocytes (CCs), as a promising donor cell source for cell-based therapy for cartilage repair, have strong tendency of hypertrophy and calcification, which limited CCs from further application in cartilage regenerative medicine. Synovium-derived stromal cells (SDSCs), have shown their beneficial effect for chondrocytes to maintain phenotype. This study aims to investigate whether SDSCs could help CCs to maintain chondrogenic phenotype and suppress hypertrophic differentiation in cartilage repairs. METHODS: CCs were directly cocultured with SDSCs in pellet or indirectly cocultured using a conditioned medium in vitro for 3 weeks. Cartilage matrix formation and hypertrophic differentiation of CCs were analyzed by RT-PCR, biochemical assays, and histological staining. Cocultured pellets were implanted into the osteochondral defects made on the femoral groove of the rats. Then, macroscopic and histological evaluations were performed. RESULTS: Pellets formed by CCs alone and CCs cocultured with SDSCs reveal equal cartilage matrix deposition. However, the gene expression of type X collagen was significantly downregulated in cocultured pellets. Immunohistochemistry analysis revealed suppressed expression of type X collagen in cocultured pellets, indicating SDSCs may suppress hypertrophic differentiation of chondrocytes. Further in indirect coculture experiment, SDSCs suppressed type X collagen expression as well and promoted the proliferation of CCs, indicating SDSCs may influence CCs by paracrine mechanism. The pellets implanted in the osteochondral defects showed good restoration effects, whereas the grafts constructed with CCs and SDSCs showed lower type X expression levels. CONCLUSION: These results suggest that SDSCs may maintain the phenotype of CCs and prevent the hypertrophic differentiation of CCs in cartilage repair.The Translational Potential of this Article: CCs is a promising donor cell source for cell-based therapy for cartilage repair. Based on our study, cocultured with SDSCs weakened the tendency of hypertrophy and calcification of CCs, which provide a potential usage of SDSCs in CCs-based cartilage repair therapy to suppress newly formed cartilage calcification and improve clinical outcomes.

10.
JBJS Case Connect ; 12(4)2022 10 01.
Article in English | MEDLINE | ID: mdl-36732040

ABSTRACT

CASE: We describe a patient with advanced Kienböck's disease, treated with 3-dimensional (3D) printing assisted costochondral transplantation. Cartilage shaping was achieved according to a biomimetic 3D-printed prosthesis designed by mirror symmetry of the healthy wrist. The inserted cartilage spacer was fixed using the autologous palmar longus tendon. After 14 months of follow-up, the patient had significant pain relief and had recovered nearly the full range of wrist motion. No significant absorption or osseous metaplasia of the cartilage was seen on the radiographic and magnetic resonance images. CONCLUSION: This costochondral transplantation strategy may offer a feasible treatment option for patients with severe Kienböck's disease.


Subject(s)
Osteonecrosis , Humans , Osteonecrosis/diagnostic imaging , Osteonecrosis/surgery , Wrist Joint/pathology , Magnetic Resonance Imaging , Prostheses and Implants , Printing, Three-Dimensional
11.
Adv Healthc Mater ; 10(18): e2100626, 2021 09.
Article in English | MEDLINE | ID: mdl-34263563

ABSTRACT

This study develops a novel strategy for regenerative therapy of musculoskeletal soft tissue defects using a dual-phase multifunctional injectable gelatin-hydroxyphenyl propionic acid (Gtn-HPA) composite. The dual-phase gel consists of stiff, degradation-resistant, ≈2-mm diameter spherical beads made from 8 wt% Gtn-HPA in a 2 wt% Gtn-HPA matrix. The results of a 3D migration assay show that both the cell number and migration distance in the dual-phase gel system are comparable with the 2 wt% mono-phase Gtn-HPA, but notably significantly higher than for 8 wt% mono-phase Gtn-HPA (into which few cells migrated). The results also show that the dual phase gel system has degradation resistance and a prolonged growth factor release profile comparable with 8 wt% mono-phase Gtn-HPA. In addition, the compressive modulus of the 2 wt% dual-phase gel system incorporating the 8 wt% bead phase is nearly four-fold higher than the 2 wt% mono-phase gel (5.3 ± 0.4 kPa versus 1.5 ± 0.06 kPa). This novel injectable dual-phase Gtn-HPA composite thus combines the advantages of low-concentration Gtn-HPA (cell migration) with high-concentration Gtn-HPA (stiffness, degradation resistance, slower chemical release kinetics) to facilitate effective reparative/regenerative processes in musculoskeletal soft tissue.


Subject(s)
Gelatin , Mesenchymal Stem Cells , Musculoskeletal Physiological Phenomena , Regeneration , Hydrogels , Tissue Engineering
12.
J Tissue Eng Regen Med ; 15(4): 299-321, 2021 04.
Article in English | MEDLINE | ID: mdl-33660950

ABSTRACT

Low back pain is an increasingly prevalent symptom mainly associated with intervertebral disc (IVD) degeneration. It is highly correlated with aging, as the nucleus pulposus (NP) dehydrates and annulus fibrosus fissure formatting, which finally results in the IVD herniation and related clinical symptoms. Hydrogels have been drawing increasing attention as the ideal candidates for IVD degeneration because of their unique properties such as biocompatibility, highly tunable mechanical properties, and especially the water absorption and retention ability resembling the normal NP tissue. Numerous innovative hydrogel polymers have been generated in the most recent years. This review article will first briefly describe the anatomy and pathophysiology of IVDs and current therapies with their limitations. Following that, the article introduces the hydrogel materials in the classification of their origins. Next, it reviews the recent hydrogel polymers explored for IVD regeneration and analyses what efforts have been made to overcome the existing limitations. Finally, the challenges and prospects of hydrogel-based treatments for IVD tissue are also discussed. We believe that these novel hydrogel-based strategies may shed light on new possibilities in IVD degeneration disease.


Subject(s)
Biocompatible Materials/pharmacology , Hydrogels/pharmacology , Intervertebral Disc/pathology , Animals , Clinical Trials as Topic , Humans , Intervertebral Disc/drug effects
13.
J Orthop Translat ; 21: 122-128, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32309137

ABSTRACT

Total hip arthroplasty is a common surgical technique, yet it has severe complications, such as loosening and repeated revision. Thus, hip-preserving surgical options should be considered first to treat cartilage defects in the femoral head, especially for younger patients. Current surgical options for chondral repair of the femoral head include microfracture, trapdoor procedure, transplantation of osteochondral allografts and autografts, and autologous chondrocyte implantation. Each of these techniques has unique advantages and limitations; however, none of them have been consented as the best practice for cartilage defects. In this review article, we also introduced a novel technique for repairing osteochondral defects of the femoral head using autologous costal cartilage grafts that may have good translational potential for cost-effective and safe applications. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE: This review updates current surgical options for reparing articular cartilage defects in the femoral head. We also introduce a novel technique for repairing osteochondral defects of the femoral head using autologous costal cartilage grafts.

14.
Ther Adv Musculoskelet Dis ; 11: 1759720X19877131, 2019.
Article in English | MEDLINE | ID: mdl-31579403

ABSTRACT

It is a great challenge to cure symptomatic lesions and considerable defects of hyaline cartilage due to its complex structure and poor self-repair capacity. If left untreated, unmatured degeneration will cause significant complications. Surgical intervention to repair cartilage may prevent progressive joint degeneration. A series of surgical techniques, including biological augmentation, microfracture and bone marrow stimulation, autologous chondrocyte implantation (ACI), and allogenic and autogenic chondral/osteochondral transplantation, have been used for various indications. However, the limited repairing capacity and the potential pitfalls of these techniques cannot be ignored. Increasing evidence has shown promising outcomes from ACI and cartilage transplantation. Nevertheless, the morbidity of autologous donor sites and limited resource of allogeneic bone have considerably restricted the wide application of these surgical techniques. Costal cartilage, which preserves permanent chondrocytes and the natural osteochondral junction, is an ideal candidate for the restoration of cartilage defects. Several in vitro and in vivo studies have shown good performance of costal cartilage transplantation. Although costal cartilage is a classic donor in plastic and cosmetic surgery, it is rarely used in skeletal cartilage restoration. In this review, we introduce the fundamental properties of costal cartilage and summarize costa-derived chondrocyte implantation and costal chondral/osteochondral transplantation. We will also discuss the pitfalls and pearls of costal cartilage transplantation. Costal chondral/osteochondral transplantation and costa-based chondrocytotherapy might be up-and-coming surgical techniques for recalcitrant cartilage lesions.

15.
J Foot Ankle Surg ; 58(3): 604-606, 2019 May.
Article in English | MEDLINE | ID: mdl-31047029

ABSTRACT

Achilles tendon lengthening is an important surgical procedure to manage gastrocnemius-soleus complex contracture. Because the Achilles tendon fibers twist like Manila rope and torsion varies widely, it is very difficult for any current lengthening procedure to be performed that accurately follows the rotation of the fibers; thus, irregular sliding or repeated cutting of the fibers may result. We present a patient with Achilles tendon contracture in whom the tendon was divided coronally along the twisted fibers using a stainless-steel wire before hemisection for Z-lengthening; thus, hemisection could be performed not only with minimal invasion but also accurately. After tendon lengthening, ankle function was restored to near normal. After a 15-month follow-up time, improvement of ankle function was well maintained, and no complication, such as crouch gait, was observed. We believe this was owed to accurate division of the tendon fibers before lengthening, preservation of the paratenon and deep fascial tube, and the use of Z-lengthening.


Subject(s)
Achilles Tendon/surgery , Contracture/surgery , Tenotomy/methods , Achilles Tendon/physiopathology , Contracture/physiopathology , Humans , Male , Young Adult
16.
Biomed Res Int ; 2015: 859456, 2015.
Article in English | MEDLINE | ID: mdl-26504839

ABSTRACT

Porous ceramic scaffolds with shapes matching the bone defects may result in more efficient grafting and healing than the ones with simple geometries. Using computer-assisted microstereolithography (MSTL), we have developed a novel gelcasting indirect MSTL technology and successfully fabricated two scaffolds according to CT images of rabbit femur. Negative resin molds with outer 3D dimensions conforming to the femur and an internal structure consisting of stacked meshes with uniform interconnecting struts, 0.5 mm in diameter, were fabricated by MSTL. The second mold type was designed for cortical bone formation. A ceramic slurry of beta-tricalcium phosphate (ß-TCP) with room temperature vulcanization (RTV) silicone as binder was cast into the molds. After the RTV silicone was completely cured, the composite was sintered at 1500°C for 5 h. Both gross anatomical shape and the interpenetrating internal network were preserved after sintering. Even cortical structure could be introduced into the customized scaffolds, which resulted in enhanced strength. Biocompatibility was confirmed by vital staining of rabbit bone marrow mesenchymal stromal cells cultured on the customized scaffolds for 5 days. This fabrication method could be useful for constructing bone substitutes specifically designed according to local anatomical defects.


Subject(s)
Bone and Bones/cytology , Calcium Phosphates/chemistry , Printing, Three-Dimensional , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Cells, Cultured , Humans , Image Processing, Computer-Assisted , Mesenchymal Stem Cells/cytology , Microtechnology , Rabbits , X-Ray Microtomography
17.
Cartilage ; 6(4): 241-51, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26425262

ABSTRACT

OBJECTIVE: To investigate the feasibility of repairing osteochondral defects of critical size by performing mosaicplasty using multiple sliced costal cartilage grafts, which enables repair of extensively injured knees using grafts from a single rib. DESIGN: Critical osteochondral defects were prepared on the femoral groove of skeletally mature Japanese white rabbits. Costal cartilage grafts from a single rib were harvested and sliced into multiple segments (approximately 3-5 mm in length). The defects were left untreated or repaired by performing mosaicplasty using costal cartilage grafts (with or without a longitudinal cut along the middle). At 4 and 12 weeks after transplantation, International Cartilage Repair Society macroscopic and histological grading was performed. RESULTS: The macroscopic score and visual histological score were significantly higher in the repaired groups than in the untreated group at 4 and 12 weeks after surgery. Histological continuous integration between grafted costal cartilage and host bone was observed in both repaired groups. CONCLUSIONS: The findings suggest that costal cartilage might be a useful alternative source for chondral grafting. We were able to repair large osteochondral defects by performing mosaicplasty using multiple sliced costal cartilage grafts from a single rib.

18.
J Biomed Mater Res B Appl Biomater ; 103(1): 84-91, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24764314

ABSTRACT

Media perfusion is often required to maintain cell viability within topographically complex 3-dimensional scaffold cultures. Osteoblast-seeded scaffolds for bone regeneration require robust cell proliferation and survival both within the scaffold and over the exterior for optimal osteogenic capacity. Conventional press-fitting cassettes ensure internal fluid flow through the scaffold but may restrict external flow around the scaffold, resulting in a barren (cell-free) external scaffold surface. In this study, we aimed to solve this problem by modifying the cassette structure to enhance external flow in an oscillatory perfusion culture system. Mouse osteoblast-like MC 3T3-E1 cells were seeded in porous ceramic scaffolds and incubated for 3 days either under static culture conditions or in an oscillatory perfusion bioreactor. Scaffolds were held in the bioreactor with either conventional press-fitting cassettes or cassettes with rings to separate the scaffold exterior from the internal cassette wall. The external surfaces of scaffolds maintained under static conditions were well seeded, but cells failed to grow deeply into the core, reflecting poor internal chemotransport. Alternatively, scaffolds cultured by perfusion with press-fitting cassettes had poor cell viability at the cassette-external scaffold surface interface, but cells were widely distributed within the scaffold core. Scaffolds cultured using the modified cassettes with 1 or 2 rings exhibited uniformly distributed living cells throughout the internal pores and over the entire external surface, possibly because of the improved medium flow over the scaffold surface. This modified oscillatory perfusion culture system may facilitate the production of engineered bone with superior osteogenic capacity for grafting.


Subject(s)
Bone Substitutes/chemistry , Osteoblasts/metabolism , Tissue Engineering , Tissue Scaffolds/chemistry , Animals , Cell Culture Techniques , Cell Line , Mice , Osteoblasts/cytology , Porosity
19.
Biofabrication ; 6(4): 045002, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25215543

ABSTRACT

Because patient bone defects are usually varied and complicated in geometry, it would be preferred to fabricate custom-made artificial bone grafts that are anatomically specific to individual patient defects. Using a rabbit femoral segment as a bone reconstruction model, we successfully produced customized ceramic scaffolds by stereolithography, which not only had an anatomically correct external shape according to computed tomography data but also contained an interconnecting internal network of channels designed for perfusion culture. Rabbit bone marrow stromal cells were isolated and cultured with these scaffolds using a novel oscillatory perfusion system that was stereolithographically fabricated to fit well to the unique scaffold shapes. After five days of three-dimensional culture with oscillatory perfusion, the cells attached and proliferated homogenously in the scaffolds. However, control cells inside the scaffolds cultured under static conditions were dead after prolonged in vitro culture. Cellular DNA content and alkaline phosphatase activities were significantly higher in the perfusion group versus the static group. Therefore, anatomically correct artificial bone can be successfully constructed using stereolithography and oscillatory culture technology, and could be useful for bone engraftment and defect repair.


Subject(s)
Bone and Bones/cytology , Cell Culture Techniques/instrumentation , Printing, Three-Dimensional , Tissue Engineering/instrumentation , Tissue Scaffolds , Alkaline Phosphatase/metabolism , Animals , Bone Marrow Cells , Cell Survival , DNA/analysis , Male , Perfusion , Porosity , Rabbits
20.
Int J Med Robot ; 10(3): 325-31, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24030893

ABSTRACT

BACKGROUND: Closed reduction is of great benefit for fracture healing. However, achieving this without sacrificing the reduction accuracy and exposing the surgeon and patient to excessive radiation is difficult. METHODS: A novel parachute guiding system (ParaEx System) was developed for closed reduction of fractures based on computed tomography data. The system included two counter guides with stainless tubular markers that could be attached to the unilateral external fixator. Comminuted tibial diaphyseal fracture models were used to validate the ParaEx System. RESULTS: The mean errors (and standard deviations) of residual rotational and translational deformity were 0.67° ± 0.45°, 0.92° ± 1.00°, and 0.64° ± 0.50° in rotation and 1.30 ± 1.10 mm, 1.13 ± 0.70 mm, and 0.94 ± 0.92 mm in translation about the X, Y, and Z axes of the local coordinate axes, respectively. CONCLUSIONS: The ParaEx System was useful for accurate closed reduction of fractures at low cost.


Subject(s)
Diaphyses/surgery , Fracture Fixation/methods , Surgery, Computer-Assisted/methods , Computer Simulation , Computer Systems , Equipment Design , Fracture Healing , Fractures, Bone/surgery , Humans , Reproducibility of Results , Tibia/surgery , Tibial Fractures/surgery , Tomography, X-Ray Computed/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...