Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 581
Filter
Add more filters

Publication year range
1.
Osteoarthritis Cartilage ; 29(5): 707-717, 2021 05.
Article in English | MEDLINE | ID: mdl-33609694

ABSTRACT

OBJECTIVE: While cartilaginous endplate (CEP) avulsion is a common finding in discectomy due to lumbar disc herniation, its roles in residual back and leg pain, associations with Modic changes (MCs) and endplate defects (EPD) remain unknown. DESIGN: Patients with a single-level lumbar disc herniation who underwent endoscopic discectomy were studied. On MR images, the adjacent endplates of the herniated disc were assessed for MCs and EPD. The presence of CEP avulsion was examined under endoscopic and visualized inspection. Back and leg pain were evaluated by a numeric rating scale (NRS) and the Oswestry Disability Index. Associations of CEP avulsion with adjacent MCs, EPD, and residual back and leg pain were examined. In addition, histological features of avulsed CEP were determined using gross staining and immunohistochemical methods. RESULTS: A total of 386 patients were included. CEP avulsion was found in 166 (43%) patients, and adjacent MCs and EPD were observed in 117 (30.3%) and 139 (36%) patients. The presence of CEP avulsion was associated with greater age, adjacent MCs (OR = 2.60, 95%CI [1.61-4.19]) and EPD (OR = 1.63, 95%CI [1.03-2.57]). Among the 187 patients with ≥2 years follow-up, CEP avulsion was associated with residual back pain (OR = 2.49, 95%CI [1.29-4.82]) and leg pain (OR = 2.25, 95%CI [1.04-4.84]). Histologically, the avulsed CEP was characterized by multiple defects, apparent inflammation, and nucleus invasion, as well as the upregulation of IL-1ß, caspase-1, and NLRP3 inflammasome. CONCLUSION: CEP avulsion was associated with MCs, EPD, and residual back and leg pain after discectomy, which may be attributed to NLRP3 inflammasome related inflammations.


Subject(s)
Back Pain/etiology , Cartilage/injuries , Diskectomy/adverse effects , Intervertebral Disc Displacement/surgery , Age Factors , Cartilage/diagnostic imaging , Cartilage/metabolism , Caspase 1/metabolism , Chronic Pain/etiology , Disability Evaluation , Female , Follow-Up Studies , Humans , Interleukin-1beta/metabolism , Lumbar Vertebrae , Magnetic Resonance Imaging , Male , Middle Aged , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pain Measurement , Retrospective Studies , Up-Regulation
2.
PLoS Comput Biol ; 16(6): e1007998, 2020 06.
Article in English | MEDLINE | ID: mdl-32584809

ABSTRACT

Post-traumatic osteoarthritis (PTOA) is associated with cartilage degradation, ultimately leading to disability and decrease of quality of life. Two key mechanisms have been suggested to occur in PTOA: tissue inflammation and abnormal biomechanical loading. Both mechanisms have been suggested to result in loss of cartilage proteoglycans, the source of tissue fixed charge density (FCD). In order to predict the simultaneous effect of these degrading mechanisms on FCD content, a computational model has been developed. We simulated spatial and temporal changes of FCD content in injured cartilage using a novel finite element model that incorporates (1) diffusion of the pro-inflammatory cytokine interleukin-1 into tissue, and (2) the effect of excessive levels of shear strain near chondral defects during physiologically relevant loading. Cytokine-induced biochemical cartilage explant degradation occurs near the sides, top, and lesion, consistent with the literature. In turn, biomechanically-driven FCD loss is predicted near the lesion, in accordance with experimental findings: regions near lesions showed significantly more FCD depletion compared to regions away from lesions (p<0.01). Combined biochemical and biomechanical degradation is found near the free surfaces and especially near the lesion, and the corresponding bulk FCD loss agrees with experiments. We suggest that the presence of lesions plays a role in cytokine diffusion-driven degradation, and also predisposes cartilage for further biomechanical degradation. Models considering both these cartilage degradation pathways concomitantly are promising in silico tools for predicting disease progression, recognizing lesions at high risk, simulating treatments, and ultimately optimizing treatments to postpone the development of PTOA.


Subject(s)
Biophysics , Cartilage/injuries , Cartilage/metabolism , Cytokines/metabolism , Inflammation Mediators/metabolism , Stress, Mechanical , Animals , Humans
3.
PLoS Comput Biol ; 16(10): e1008275, 2020 10.
Article in English | MEDLINE | ID: mdl-33027251

ABSTRACT

Inconsistent therapeutic efficacy of mesenchymal stem cells (MSCs) in regenerative medicine has been documented in many clinical trials. Precise prediction on the therapeutic outcome of a MSC therapy based on the patient's conditions would provide valuable references for clinicians to decide the treatment strategies. In this article, we performed a meta-analysis on MSC therapies for cartilage repair using machine learning. A small database was generated from published in vivo and clinical studies. The unique features of our neural network model in handling missing data and calculating prediction uncertainty enabled precise prediction of post-treatment cartilage repair scores with coefficient of determination of 0.637 ± 0.005. From this model, we identified defect area percentage, defect depth percentage, implantation cell number, body weight, tissue source, and the type of cartilage damage as critical properties that significant impact cartilage repair. A dosage of 17 - 25 million MSCs was found to achieve optimal cartilage repair. Further, critical thresholds at 6% and 64% of cartilage damage in area, and 22% and 56% in depth were predicted to significantly compromise on the efficacy of MSC therapy. This study, for the first time, demonstrated machine learning of patient-specific cartilage repair post MSC therapy. This approach can be applied to identify and investigate more critical properties involved in MSC-induced cartilage repair, and adapted for other clinical indications.


Subject(s)
Cartilage , Machine Learning , Mesenchymal Stem Cell Transplantation , Tissue Engineering/methods , Animals , Cartilage/cytology , Cartilage/injuries , Cartilage/surgery , Computational Biology , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Models, Biological , Rabbits , Rats , Swine
4.
J Nanobiotechnology ; 19(1): 343, 2021 Oct 26.
Article in English | MEDLINE | ID: mdl-34702302

ABSTRACT

OBJECTIVES: This study aimed to investigate the roles of adipose mesenchymal stem cell (AMSC)-derived extracellular vesicles (EVs) binding with chitosan oligosaccharides (COS) in cartilage injury, as well as the related mechanisms. RESULTS: IL-1ß treatment significantly inhibited the viability and migration of chondrocytes and enhanced cell apoptosis (P < 0.05), while chitosan oligosaccharides and extracellular vesicles-chitosan oligosaccharide conjugates (EVs-COS/EVs-COS conjugates) reversed the changes induced by IL-1ß (P < 0.05), and the effects of extracellular vesicles-chitosan oligosaccharide conjugates were better than those of chitosan oligosaccharides (P < 0.05). After cartilage damage, IL-1ß, OPN, and p53 were significantly upregulated, COL1A1, COL2A1, OCN, RUNX2, p-Akt/Akt, PI3K, c-Myc, and Bcl2 were markedly downregulated, and extracellular vesicles-chitosan oligosaccharide conjugates reversed the expression induced by cartilage injury. Through sequencing, 760 differentially expressed genes (DEGs) clustered into four expression patterns were associated with negative regulation of the canonical Wnt, PI3K-Akt, AMPK, and MAPK signaling pathways. CONCLUSION: Extracellular vesicles-chitosan oligosaccharide conjugates may serve as a new cell-free biomaterial to facilitate cartilage injury repair and improve osteoarthritis.


Subject(s)
Cartilage , Chitosan , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Animals , Cartilage/drug effects , Cartilage/injuries , Cartilage/metabolism , Cells, Cultured , Chitosan/chemistry , Chitosan/pharmacology , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/metabolism , Extracellular Vesicles/chemistry , Female , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Oligosaccharides/chemistry , Oligosaccharides/pharmacology , Osteoarthritis/metabolism , Rats , Rats, Wistar , Specific Pathogen-Free Organisms , Transcriptome/genetics , Wound Healing/drug effects
5.
J Cell Physiol ; 235(2): 979-992, 2020 02.
Article in English | MEDLINE | ID: mdl-31267533

ABSTRACT

This study is carried out to investigate the role of microRNA-26a (miR-26a) in cartilage injury and chondrocyte proliferation and apoptosis in rats with rheumatoid arthritis (RA) by regulating expression of CTGF. A rat model of RA induced by type II collagen was established. The rats were assigned into normal, RA, RA + mimics negative control (NC), and RA + miR-26a mimics groups, and the cells were classified into blank, mimics NC, and miR-26a mimics groups. The degree of secondary joint swelling and arthritis index, expression of miR-26a, pathological changes, proliferation and apoptosis of chondrocytes, and expression of CTGF, interleukin-1ß (IL-1ß), IL-6, tumor necrosis factor-α, Bax, and Bcl-2 were also determined through a series of experiments. The targeting relationship between miR-26a and CTGF was verified. Initially, downregulated miR-26a was found in cartilage tissues and inflammatory articular chondrocytes of RA rats. In addition, CTGF was determined as a direct target gene of miR-26a, and upregulation of miR-26a inhibited CTGF expression in cartilage tissues of RA rats. Furthermore, upregulation of miR-26a reduced swelling and inflammation of joints, inhibited cartilage damage, apoptosis of chondrocytes, inflammatory injury, promotes proliferation, and inhibited apoptosis of inflammatory articular chondrocytes, which may be correlated with the targeting inhibition of CTGF expression. Collectively, the results demonstrate that upregulating the expression of miR-26a could attenuate cartilage injury, stimulate the proliferation, and inhibit apoptosis of chondrocytes in RA rats.


Subject(s)
Arthritis, Rheumatoid/chemically induced , Cartilage/injuries , Cell Proliferation/physiology , Chondrocytes/metabolism , Connective Tissue Growth Factor/metabolism , MicroRNAs/metabolism , Animals , Apoptosis/physiology , Cartilage/metabolism , Collagen Type II/toxicity , Connective Tissue Growth Factor/genetics , Gene Expression Regulation , MicroRNAs/genetics , Rats , Rats, Sprague-Dawley
6.
Connect Tissue Res ; 61(2): 229-243, 2020 03.
Article in English | MEDLINE | ID: mdl-31134817

ABSTRACT

Purpose: Scaffold-free cartilage tissue engineering circumvents issues with scaffold seeding, potential toxicity response, and impaired host integration. However, precisely controlling and maintaining a scaffold-free construct shape have been challenging. We explored the feasibility of microneedle arrays to print tissue using cellular microspheroids as building blocks.Materials and Methods: Human embryonic-derived mesenchymal stem cells or infrapatellar fat pad mesenchymal stem cells were used to create microspheroids of 500 µm in diameter, which were assembled on microneedle arrays in a predefined arrangement using a robotic system under computer vision. Microspheroids on microneedles were cultured to permit fusion into a tissue construct. Infrapatellar fat pad mesenchymal stem cell constructs were either implanted into chondral defects created in human osteoarthritic cartilage explants or maintained on the microneedle array for 3 weeks. Embryonic-derived mesenchymal stem cell constructs were designed to be press-fit into 3 mm subchondral defects in New Zealand White rabbits and maintained for up to 8 weeks to assess retention, early tissue repair, and more mature cartilage regeneration.Results: Microspheroids of both cell types fused together in culture to form neotissues of predefined shape and size. Infrapatellar fat pad mesenchymal stem cell neotissues expressed high levels of chondrogenic genes and integrated with the surrounding osteoarthritic host cartilage. Embryonic-derived mesenchymal stem cell constructs generated chondrogenic neotissue in vivo as early as 2 weeks and more mature tissue by 8 weeks with increased glycosaminoglycan deposition.Conclusions: We constructed defined scaffold-free shapes by bioprinting and fusing microspheroids. Proof of concept was shown in the repair of ex vivo osteoarthritic human cartilage and in vivo rabbit osteochondral (OC) defects.


Subject(s)
Cartilage , Chondrogenesis , Human Embryonic Stem Cells/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Osteoarthritis , Robotic Surgical Procedures , Tissue Engineering , Aged , Animals , Cartilage/injuries , Cartilage/metabolism , Cartilage/pathology , Female , Human Embryonic Stem Cells/pathology , Humans , Male , Mesenchymal Stem Cells/pathology , Middle Aged , Needles , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteoarthritis/therapy , Rabbits
7.
FASEB J ; 32(10): 5298-5311, 2018 10.
Article in English | MEDLINE | ID: mdl-29688810

ABSTRACT

The continuous presence of TGF-ß is critically important to induce effective chondrogenesis. To investigate chondrogenesis in a cartilage defect, we tested the hypothesis that the implantation of TGF-ß1-releasing scaffolds improves very early cartilage repair in vivo. Spatiotemporal controlled release of TGF-ß1 was achieved from multiblock scaffolds that were implanted in osteochondral defects in the medial femoral condyles of adult minipigs. We observed a sustained presence of TGF-ß1 at 4 wk in vivo, which significantly promoted structural aspects of early overall cartilage repair, especially cellularity, cellular morphology, and safranin O staining intensity. Furthermore, early aggrecan and type II collagen production were both increased in specific topographic patterns in cartilaginous repair tissue. Sustained release of TGF-ß1 also increased cell numbers and proliferation, staining intensities for the stem cell surface marker, CD105, and number of stromal cell-derived factor-1 (SDF-1) -positive cells within cartilaginous repair tissue. These data identify a mechanism by which TGF-ß1 modulates early chondrogenesis by primarily increasing the number of progenitor cells arising from the subchondral bone marrow compartment via the SDF-1/chemokine (CXC motif) receptor 4 pathway, their proliferation, differentiation, and extracellular matrix deposition in specific topographic patterns, highlighting the pivotal role played by TGF-ß1 during this crucial phase.-Asen, A.-K., Goebel, L., Rey-Rico, A., Sohier, J., Zurakowski, D., Cucchiarini, M., Madry, H. Sustained spatiotemporal release of TGF-ß1 confers enhanced very early chondrogenic differentiation during osteochondral repair in specific topographic patterns.


Subject(s)
Cartilage , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chondrogenesis/drug effects , Transforming Growth Factor beta , Animals , Cartilage/injuries , Cartilage/metabolism , Cartilage/physiology , Chemokine CXCL12/metabolism , Drug Implants , Endoglin/metabolism , Receptors, CXCR4/metabolism , Swine , Swine, Miniature , Transforming Growth Factor beta/pharmacokinetics , Transforming Growth Factor beta/pharmacology
8.
Connect Tissue Res ; 60(6): 597-610, 2019 11.
Article in English | MEDLINE | ID: mdl-31020864

ABSTRACT

Purpose: There is a clinical need to better characterize tissue sources being used for stem cell therapies. This study focuses on comparison of cells and connective tissue progenitors (CTPs) derived from native human infrapatellar fatpad (IPFP), synovium (SYN), and periosteum (PERI). Materials and Methods: IPFP, SYN, PERI were harvested from twenty-eight patients undergoing arthroplasty. CTPs were quantitatively characterized using automated colony-forming-unit assay to compare total nucleated cell concentration-[Cell], cells/mg; prevalence-(PCTP), CTPs/million nucleated cells; CTP concentration-[CTP], CTPs/mg; proliferation and differentiation potential; and correlate outcomes with patient's age and gender. Results: [Cell] did not differ between IPFP, SYN, and PERI. PCTP was influenced by age and gender: patients >60 years, IPFP and SYN had higher PCTP than PERI (p < 0.001) and females had higher PCTP in IPFP (p < 0.001) and SYN (p = 0.001) than PERI. [CTP] was influenced by age: patients <50 years, SYN (p = 0.0165) and PERI (p < 0.001) had higher [CTP] than IPFP; patients between 60 and 69 years, SYN (p < 0.001) had higher [CTP] than PERI; patients >70 years, IPFP (p = 0.006) had higher [CTP] than PERI. In patients >60 years, proliferation potential of CTPs differed significantly (SYN>IPFP>PERI); however, differentiation potentials were comparable between all three tissue sources. Conclusion: SYN and IPFP may serve as a preferred tissue source for patients >60 years, and PERI along with SYN and IPFP may serve as a preferred tissue source for patients <60 years for cartilage repair. However, the heterogeneity among the CTPs in any given tissue source suggests performance-based selection might be useful to optimize cell-sourcing strategies to improve efficacy of cellular therapies for cartilage repair.


Subject(s)
Adipose Tissue/metabolism , Chondrogenesis , Patella/metabolism , Periosteum/metabolism , Stem Cells/metabolism , Synovial Membrane/metabolism , Adipose Tissue/pathology , Adult , Aged , Aged, 80 and over , Cartilage/injuries , Cartilage/metabolism , Cartilage/pathology , Cell- and Tissue-Based Therapy , Female , Humans , Male , Middle Aged , Patella/pathology , Periosteum/pathology , Stem Cells/pathology , Synovial Membrane/pathology
9.
Eur Arch Otorhinolaryngol ; 276(7): 1981-1986, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30937560

ABSTRACT

INTRODUCTION: Septoplasty is one of the most common otolaryngologic procedures. Previous studies have reported that the overall rate of significant change in cosmetic appearance of the nose after septoplasty ranged from 0.4 to 3.4%, and saddle nose was the most commonly cited deformity. In this study, we evaluated the risk factors for intraoperative saddle nose in a group of septoplasty patients. METHODS: This case-control study (1:2 case:control) was conducted based on retrospective chart review. Intraoperative saddle nose was observed in 108 (5.1%) of 2106 patients who underwent septoplasty in our center between January 2008 and December 2017. The control group consisted of 216 randomly selected, hospital-matched septoplasty patients who had no intraoperative saddle nose deformity in the same period. The demographic data, preoperative endoscopic findings, and surgical procedures of the two groups were analyzed to identify possible risk factors of intraoperative saddle nose deformity. RESULTS: The mean ages of the two groups were 34.8 years (saddle group) and 33.2 years (control group). In multivariate logistic regression analysis, clinical risk factors associated with intraoperative saddle nose were female gender (OR 3.39; 95% CI 1.76-6.54; p < 0.01), severe caudal septal deviation (OR 2.22; 95% CI 1.30-3.79; p = 0.003), and intraoperative finding of septal cartilage fracture (OR 3.96; 95% CI 1.92-8.19; p < 0.01). CONCLUSIONS: Severe caudal septal deviation, intraoperative fracture of septal cartilage, and female gender were risk factors for intraoperative saddle nose deformity in our study population.


Subject(s)
Intraoperative Complications , Nasal Septum/surgery , Nose Deformities, Acquired , Rhinoplasty/adverse effects , Adult , Cartilage/injuries , Case-Control Studies , Female , Humans , Intraoperative Complications/diagnosis , Intraoperative Complications/epidemiology , Male , Middle Aged , Nose Deformities, Acquired/diagnosis , Nose Deformities, Acquired/epidemiology , Nose Deformities, Acquired/etiology , Nose Diseases/surgery , Republic of Korea , Retrospective Studies , Rhinoplasty/methods , Risk Assessment , Risk Factors
10.
Int J Mol Sci ; 20(24)2019 Dec 13.
Article in English | MEDLINE | ID: mdl-31847077

ABSTRACT

The unavailability of sufficient numbers of human primary cells is a major roadblock for in vitro repair of bone and/or cartilage, and for performing disease modelling experiments. Immortalized mesenchymal stromal cells (iMSCs) may be employed as a research tool for avoiding these problems. The purpose of this review was to revise the available literature on the characteristics of the iMSC lines, paying special attention to the maintenance of the phenotype of the primary cells from which they were derived, and whether they are effectively useful for in vitro disease modeling and cell therapy purposes. This review was performed by searching on Web of Science, Scopus, and PubMed databases from 1 January 2015 to 30 September 2019. The keywords used were ALL = (mesenchymal AND ("cell line" OR immortal*) AND (cartilage OR chondrogenesis OR bone OR osteogenesis) AND human). Only original research studies in which a human iMSC line was employed for osteogenesis or chondrogenesis experiments were included. After describing the success of the immortalization protocol, we focused on the iMSCs maintenance of the parental phenotype and multipotency. According to the literature revised, it seems that the maintenance of these characteristics is not guaranteed by immortalization, and that careful selection and validation of clones with particular characteristics is necessary for taking advantage of the full potential of iMSC to be employed in bone and cartilage-related research.


Subject(s)
Bone Regeneration , Bone and Bones , Cartilage , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Bone and Bones/injuries , Bone and Bones/metabolism , Bone and Bones/pathology , Cartilage/injuries , Cartilage/metabolism , Cartilage/pathology , Chondrogenesis , Humans , Mesenchymal Stem Cells/pathology , Osteogenesis
11.
Int J Mol Sci ; 20(12)2019 Jun 14.
Article in English | MEDLINE | ID: mdl-31207966

ABSTRACT

Joint injuries are highly associated with the development of post-traumatic osteoarthritis. Previous studies revealed cell- and matrix-protective effects of N-acetylcysteine (NAC) after ex vivo cartilage trauma, while chondroanabolic stimulation with bone morphogenetic protein 7 (BMP7) enhanced type II collagen (COL2) expression. Here, as a next step, we investigated the combined and individual efficacy of intra-articular antioxidative and chondroanabolic treatment in a rabbit in vivo cartilage trauma model. Animals were randomly divided into group A (right joint: trauma (T); left joint: T+BMP7) and group B (right joint: T+NAC; left joint: T+BMP7+NAC). Condyles were impacted with the use of a spring-loaded impact device to ensure defined, single trauma administration. After 12 weeks, histopathological analysis was performed and the presence of matrix metalloproteinase 13 (MMP-13) and COL2 was assessed. Trauma-induced hypocellularity, MMP-13 expression, and cell cluster formation were reduced in NAC-treated animals. In contrast, BMP7 further increased cluster formation. Moreover, synovial concentrations of COL2 carboxy propeptide (CPII) and proteoglycan staining intensities were enhanced in NAC- and NAC+BMP7-treated joints. For the first time, the efficacy of NAC regarding early harm reduction after blunt cartilage trauma was demonstrated in vivo. However, parallel administration of BMP7 was not significantly superior compared to NAC alone.


Subject(s)
Acetylcysteine/therapeutic use , Cartilage/metabolism , Osteoarthritis/drug therapy , Regeneration , Wounds, Nonpenetrating/complications , Acetylcysteine/pharmacology , Animals , Bone Morphogenetic Protein 7/pharmacology , Bone Morphogenetic Protein 7/therapeutic use , Cartilage/drug effects , Cartilage/injuries , Cartilage/physiology , Collagen Type II/metabolism , Female , Matrix Metalloproteinase 13/metabolism , Osteoarthritis/etiology , Rabbits , Wounds, Nonpenetrating/drug therapy
12.
J Magn Reson Imaging ; 47(1): 69-77, 2018 01.
Article in English | MEDLINE | ID: mdl-28449249

ABSTRACT

PURPOSE: To quantitatively evaluate the cartilage alteration of talus for chronic lateral ankle instability (LAI) with isolated anterior talofibular ligament (ATFL) tear and combined ATFL and calcaneofibular ligament (CFL) tear using T2 -mapping at 3.0T. MATERIALS AND METHODS: In all, 27 patients including 17 with isolated ATFL tear and 10 with ATFL+CFL tear, and 21 healthy subjects were recruited. All participants underwent T2 -mapping scan at 3T and patients completed American Orthopaedic Foot and Ankle Society (AOFAS) scoring. The total talar cartilage (TTC) was segmented into six compartments: medial anterior (MA), medial center (MC), medial posterior (MP), lateral anterior (LA), lateral center (LC), and lateral posterior (LP). The T2 value of each compartment was measured from T2 -mapping images. Data were analyzed with one-way analysis of variance (ANOVA), Student's t-test, and Pearson's correlation coefficient. RESULTS: The T2 values of MA, MC, MP, TTC in the ATFL group and MA, MC, MP, LC, LP, TTC in the ATFL+CFL group were higher than those in the control group (P < 0.05). Moreover, the T2 values of MC, MP, LC, and TTC in the ATFL+CFL group were higher than those in the ATFL group (P < 0.05). The T2 values of MA in both patient groups were negatively correlated with AOFAS scores (r = -0.596, r = -0.690, P < 0.05). CONCLUSION: Chronic LAI with ATFL tear had a trend of increasing cartilage T2 values in talar trochlea, mainly involving medial cartilage compartments. Chronic LAI with ATFL+CFL tear might result in higher T2 values in a much larger cartilage region than with ATFL tear. MA could be the main cartilage compartment that may affect the patient's clinical symptoms. LEVEL OF EVIDENCE: 1 Technical Efficacy: Stage 3 J. Magn. Reson. Imaging 2018;47:69-77.


Subject(s)
Ankle Joint/diagnostic imaging , Ankle/diagnostic imaging , Joint Instability/diagnostic imaging , Lateral Ligament, Ankle/diagnostic imaging , Magnetic Resonance Imaging , Adult , Ankle Injuries/diagnostic imaging , Cartilage/diagnostic imaging , Cartilage/injuries , Disease Progression , Female , Healthy Volunteers , Humans , Lateral Ligament, Ankle/injuries , Male , Observer Variation , Talus , Young Adult
13.
Inflamm Res ; 67(1): 5-20, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28803264

ABSTRACT

INTRODUCTION: Our knowledge of extracellular matrix (ECM) structure and function has increased enormously over the last decade or so. There is evidence demonstrating that ECM provides signals affecting cell adhesion, shape, migration, proliferation, survival, and differentiation. ECM presents many domains that become active after proteolytic cleavage. These active ECM fragments are called matrikines which play different roles; in particular, they may act as potent inflammatory mediators during cartilage injury. FINDINGS: A major component of the ECM that undergoes dynamic regulation during cartilage damage and inflammation is the non-sulphated glycosaminoglycan (GAG) hyaluronan (HA). In this contest, HA is the most studied because of its different activity due to the different polymerization state. In vivo evidences have shown that low molecular weight HA exerts pro-inflammatory action, while high molecular weight HA possesses anti-inflammatory properties. Therefore, the beneficial HA effects on arthritis are not only limited to its viscosity and lubricant action on the joints, but it is especially due to a specific and effective anti-inflammatory activity. Several in vitro experimental investigations demonstrated that HA treatment may regulate different biochemical pathways involved during the cartilage damage. Emerging reports are suggesting that the ability to recognize receptors both for the HA degraded fragments, whether for the high-polymerized native HA involve interaction with integrins, toll-like receptors (TLRs), and the cluster determinant (CD44). The activation of these receptors induced by small HA fragments, via the nuclear factor kappa-light-chain enhancer of activated B cell (NF-kB) mediation, directly or other different pathways, produces the transcription of a large number of damaging intermediates that lead to cartilage erosion. CONCLUSIONS: This review briefly summarizes a number of findings of the recent studies focused on the protective effects of HA, at the different polymerization states, on experimental arthritis in vitro both in animal and human cultured chondrocytes.


Subject(s)
Cartilage/injuries , Chondrocytes/drug effects , Hyaluronic Acid/pharmacology , Protective Agents/pharmacology , Animals , Humans
14.
Adv Exp Med Biol ; 1058: 53-75, 2018.
Article in English | MEDLINE | ID: mdl-29691817

ABSTRACT

Considerable advances in tissue engineering and regeneration have been accomplished over the last decade. Bioceramics have been developed to repair, reconstruct, and substitute diseased parts of the body and to promote tissue healing as an alternative to metallic implants. Applications embrace hip, knee, and ligament repair and replacement, maxillofacial reconstruction and augmentation, spinal fusion, bone filler, and repair of periodontal diseases. Bioceramics are well-known for their superior wear resistance, high stiffness, resistance to oxidation, and low coefficient of friction. These specially designed biomaterials are grouped in natural bioceramics (e.g., coral-derived apatites), and synthetic bioceramics, namely bioinert ceramics (e.g., alumina and zirconia), bioactive glasses and glass ceramics, and bioresorbable calcium phosphates-based materials. Physicochemical, mechanical, and biological properties, as well as bioceramics applications in diverse fields of tissue engineering are presented herein. Ongoing clinical trials using bioceramics in osteochondral tissue are also considered. Based on the stringent requirements for clinical applications, prospects for the development of advanced functional bioceramics for tissue engineering are highlighted for the future.


Subject(s)
Bone Regeneration , Bone and Bones , Cartilage , Ceramics/chemistry , Tissue Engineering/methods , Animals , Bone and Bones/injuries , Bone and Bones/metabolism , Bone and Bones/pathology , Cartilage/injuries , Cartilage/metabolism , Cartilage/pathology , Humans , Regenerative Medicine/methods
15.
Adv Exp Med Biol ; 1058: 265-278, 2018.
Article in English | MEDLINE | ID: mdl-29691826

ABSTRACT

Platelet lysates (PLs) are a natural source of growth factors (GFs) known for its stimulatory role on stem cells which can be obtained after activation of platelets from blood plasma. The possibility to use PLs as growth factor source for tissue healing and regeneration has been pursued following different strategies. Platelet lysates are an enriched pool of growth factors which can be used as either a GFs source or as a three-dimensional (3D) hydrogel. However, most of current PLs-based hydrogels lack stability, exhibiting significant shrinking behavior. This chapter focuses on the application of supercritical fluid technology to develop three-dimensional architectures of PL constructs, crosslinked with genipin. The proposed technology allows in a single step operation the development of mechanically stable porous structures, through chemical crosslinking of the growth factors present in the PL pool, followed by supercritical drying of the samples. Furthermore gradient structures of PL-based structures with bioactive glass are also presented and are described as an interesting approach to the treatment of osteochondral defects.


Subject(s)
Blood Platelets/chemistry , Bone and Bones , Cartilage , Cross-Linking Reagents/chemistry , Hydrogels/chemistry , Intercellular Signaling Peptides and Proteins/chemistry , Iridoids/chemistry , Animals , Bone and Bones/injuries , Bone and Bones/metabolism , Bone and Bones/pathology , Cartilage/injuries , Cartilage/metabolism , Cartilage/pathology , Humans , Porosity
16.
Adv Exp Med Biol ; 1058: 281-304, 2018.
Article in English | MEDLINE | ID: mdl-29691827

ABSTRACT

Gellan gum (GG) is a widely explored natural polysaccharide that has been gaining attention in tissue engineering (TE) and regenerative medicine field, and more recently in osteochondral TE approaches. Taking advantage of its inherent features such as biocompatibility, biodegradability, similarity with the extracellular matrix and easy functionalization, GG-based hydrogels have been studied for their potential for cartilage and bone tissue regeneration. Several preclinical studies describe the successful outcome of GG in cartilage tissue engineering. By its turn, GG composites have also been proposed in several strategies to guide bone formation. The big challenge in osteochondral TE approaches is still to achieve cartilage and bone regeneration simultaneously through a unique integrated bifunctional construct. The potential of GG to be used as polymeric support to reach both bone and cartilage regeneration has been demonstrated. This chapter provides an overview of GG properties and the functionalization strategies employed to tailor its behaviour to a particular application. The use of GG in soft and hard tissues regeneration approaches, as well in osteochondral integrated TE strategies is also revised.


Subject(s)
Bone Regeneration , Bone and Bones , Cartilage , Hydrogels/chemistry , Polysaccharides, Bacterial/chemistry , Regenerative Medicine/methods , Tissue Engineering/methods , Animals , Bone and Bones/chemistry , Bone and Bones/injuries , Bone and Bones/metabolism , Bone and Bones/pathology , Cartilage/chemistry , Cartilage/injuries , Cartilage/metabolism , Cartilage/pathology , Humans
17.
Adv Exp Med Biol ; 1058: 305-325, 2018.
Article in English | MEDLINE | ID: mdl-29691828

ABSTRACT

Osteochondral lesions treatment and regeneration demands biomimetic strategies aiming physicochemical and biological properties of both bone and cartilage tissues, with long-term clinical outcomes. Hydrogels and scaffolds appeared as assertive approaches to guide the development and structure of the new osteochondral engineered tissue. Moreover, these structures alone or in combination with cells and bioactive molecules bring the mechanical support after in vitro and in vivo implantation. Moreover, multilayered structures designed with continuous interfaces furnish appropriate features of the cartilage and subchondral regions, namely microstructure, composition, and mechanical properties. Owing the potential as scaffolding materials, natural and synthetic polymers, bioceramics, and composites have been employed. Particularly, significance is attributed to the natural-based biopolymer silk fibroin from the Bombyx mori silkworm, considering its unique mechanical and biological properties. The significant studies on silk fibroin-based structures, namely hydrogels and scaffolds, towards bone, cartilage, and osteochondral tissue repair and regeneration are overviewed herein. The developed biomimetic strategies, processing methodologies, and final properties of the structures are summarized and discussed in depth.


Subject(s)
Bone Regeneration , Bone and Bones , Cartilage , Fibroins/chemistry , Hydrogels/chemistry , Tissue Scaffolds/chemistry , Animals , Bombyx , Bone and Bones/injuries , Bone and Bones/metabolism , Bone and Bones/pathology , Cartilage/injuries , Cartilage/metabolism , Cartilage/pathology , Humans
18.
Adv Exp Med Biol ; 1058: 327-355, 2018.
Article in English | MEDLINE | ID: mdl-29691829

ABSTRACT

Injectable hydrogels have demonstrated being a promising strategy for cartilage and bone tissue engineering applications, owing to their minimal invasive injection procedure, easy incorporation of cells and bioactive molecules, improved contact with the surrounding tissues and ability to match defects with complex irregular shapes, characteristics of osteoarthritic pathology. These unique properties make them highly suitable bioscaffolds for treating defects which are otherwise not easily accessible without and invasive surgical procedure. In this book chapter it has been summarized the novel appropriate injectable hydrogels for cartilage and bone tissue engineering applications of the last few years, including the most commonly used materials for the preparation, both natural and synthetic, and their fabrication techniques. The design of a suitable injectable hydrogel with an adequate gelation time that gathers perfect bioactive, biocompatible, biodegradable and good mechanical properties for clinical repair of damaged cartilage and bone tissue is a challenge of significant medical interest that remain to be achieved.


Subject(s)
Biocompatible Materials/chemistry , Bone Regeneration , Bone and Bones , Cartilage , Cross-Linking Reagents/chemistry , Hydrogels/chemistry , Tissue Scaffolds/chemistry , Animals , Bone and Bones/injuries , Bone and Bones/metabolism , Bone and Bones/pathology , Cartilage/injuries , Cartilage/metabolism , Cartilage/pathology , Humans
19.
Adv Exp Med Biol ; 1058: 391-413, 2018.
Article in English | MEDLINE | ID: mdl-29691832

ABSTRACT

Osteochondral lesions are frequent and important causes of pain and disability. These lesions are induced by traumatic injuries or by diseases that affect both the cartilage surface and the subchondral bone. Due to the limited cartilage ability to regenerate and self-repair, these lesions tend to gradually worsen and progress towards osteoarthritis. The clinical, social, and economic impact of the osteochondral lesions is impressive and although therapeutic alternatives are under discussion, a consensus is not yet been achieved. Over the previous decade, new strategies based on innovative tissue engineering approaches have been developed with promising results. However, in order those products reach the market and help the actual patient in an effective manner, there is still a lot of work to be done. The current state of the implications, clinical aspects, and available treatments for this pathology, as well as the ongoing preclinical and clinical trials are presented in this chapter.


Subject(s)
Bone and Bones , Cartilage , Osteoarthritis , Tissue Engineering/methods , Animals , Bone and Bones/injuries , Bone and Bones/metabolism , Bone and Bones/pathology , Cartilage/injuries , Cartilage/metabolism , Cartilage/pathology , Clinical Trials as Topic , Humans , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteoarthritis/therapy
20.
Adv Exp Med Biol ; 1058: 359-372, 2018.
Article in English | MEDLINE | ID: mdl-29691830

ABSTRACT

Mesenchymal stem cells (MSCs) are pluripotent stem cells with the ability to differentiate into a variety of other connective tissue cells, such as chondral, bony, muscular, and tendon tissue. Bone marrow-derived MSCs are pluripotent cells that can differentiate among others into osteoblasts, adipocytes and chondrocytes.Bone marrow-derived cells may represent the future in osteochondral repair. A one-step arthroscopic technique is developed for cartilage repair, using a device to concentrate bone marrow-derived cells and collagen powder or hyaluronic acid membrane as scaffolds for cell support and platelet gel.The rationale of the "one-step technique" is to transplant the entire bone-marrow cellular pool instead of isolated and expanded mesenchymal stem cells allowing cells to be processed directly in the operating room, without the need for a laboratory phase. For an entirely arthroscopic implantation are employed a scaffold and the instrumentation previously applied for ACI; in addition to these devices, autologous platelet-rich fibrin (PRF) is added in order to provide a supplement of growth factors. Results of this technique are encouraging at mid-term although long-term follow-up is still needed.


Subject(s)
Bone Marrow Cells , Bone and Bones , Cartilage , Cells, Immobilized , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells , Tissue Engineering/methods , Adipocytes/metabolism , Adipocytes/pathology , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone and Bones/injuries , Bone and Bones/metabolism , Bone and Bones/pathology , Cartilage/injuries , Cartilage/metabolism , Cartilage/pathology , Cell Differentiation , Cells, Immobilized/metabolism , Cells, Immobilized/pathology , Cells, Immobilized/transplantation , Chondrocytes/metabolism , Chondrocytes/pathology , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Osteoblasts/metabolism , Osteoblasts/pathology
SELECTION OF CITATIONS
SEARCH DETAIL