101
|
Nowicki M, Castro NJ, Rao R, Plesniak M, Zhang LG. Integrating three-dimensional printing and nanotechnology for musculoskeletal regeneration. NANOTECHNOLOGY 2017; 28:382001. [PMID: 28762957 PMCID: PMC5612478 DOI: 10.1088/1361-6528/aa8351] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
The field of tissue engineering is advancing steadily, partly due to advancements in rapid prototyping technology. Even with increasing focus, successful complex tissue regeneration of vascularized bone, cartilage and the osteochondral interface remains largely illusive. This review examines current three-dimensional printing techniques and their application towards bone, cartilage and osteochondral regeneration. The importance of, and benefit to, nanomaterial integration is also highlighted with recent published examples. Early-stage successes and challenges of recent studies are discussed, with an outlook to future research in the related areas.
Collapse
Affiliation(s)
- Margaret Nowicki
- Department of Mechanical and Aerospace Engineering, The George Washington University, 800 22nd Street, NW, Washington DC 20052, United States of America
| | | | | | | | | |
Collapse
|
102
|
Abstract
BACKGROUND Scapholunate ligament injury is a commonly occurring carpal ligament injury. Pathology associated with scapholunate ligament injury depends on several factors such as the time after injury, type of injury (instability) and the development of osteoarthritis. The aim of this study was to investigate and compare contact mechanics in the lunocapitate and scaphocapitate joints in the normal, injured (scapholunate dissociation) and repaired (postoperative) wrist. METHODS Four human subjects with scapholunate ligament dissociation participated in this study. MR images of normal (contralateral), injured and postoperative wrists were obtained during relaxed condition and during active light grasp. Relaxed MR images were used to construct model geometry (bones with cartilage) for the capitate, lunate and scaphoid. Kinematic transformations were obtained by using image registration between the unloaded and functionally loaded image sets. Joint surface contact mechanics were then calculated. FINDINGS All contact measures (contact force, pressure, mean pressure and area) tended to increase with injury in both articulations. A significantly higher contact area was found in the injured scaphocapitate joint compared to normal. A significant increase in peak pressure was observed in the postoperative state compared to normal. INTERPRETATION Injury to the scapholunate ligament increased contact measures, suggesting a risk for onset of osteoarthritis in both the scaphocapitate and lunocapitate joints. Surgical repair appeared to restore most measures of contact mechanics to near normal values, more so for the lunocapitate joint when compared to scaphocapitate joint. The elevated postoperative peak pressures indicate the difficulty to fully restore joint mechanics.
Collapse
|
103
|
Li B, Bai L, Shen P, Sun Y, Chen Z, Wen Y. Identification of differentially expressed microRNAs in knee anterior cruciate ligament tissues surgically removed from patients with osteoarthritis. Int J Mol Med 2017; 40:1105-1113. [PMID: 28765881 PMCID: PMC5593459 DOI: 10.3892/ijmm.2017.3086] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 07/17/2017] [Indexed: 01/12/2023] Open
Abstract
The degradation of cruciate ligaments is frequently observed in degenerative joint diseases, such as osteoarthritis (OA). The present study aimed to identify the differentially expressed microRNAs (miRNAs or miRs) in knee anterior cruciate ligament (ACL) tissues derived from patients with OA and in health subjects (non-OA). By using Affymetrix miRNA 4.0 microarrays, a total of 22 miRNAs (including let-7f-5p, miR-26b-5p and miR-146a-5p) were found to be upregulated, while 17 (including miR-18a-3p, miR-138-5p and miR-485-3p) were downregulated in the osteoarthritic ACL tissues (fold change ≥2, P-value <0.05). The expression levels of 12 miRNAs were validated by quantitative PCR, and the corresponding results revealed an excellent correlation with the microarray data (R2=0.889). Genes (such as a disintegrin and metalloproteinase domain with thrombospondin type-1 motifs, bone morphogenetic protein-2, runt related transcription factor-2, collagen-1A1 and 2, interleukin-6 and transforming growth factor-β) involved in cartilage development and remodeling, collagen biosynthesis and degradation, inflammatory response and extracellular matrix homeostasis were predicted as potential targets of the dysregulated miRNAs. Moreover, a large set of putative genes were enriched in OA pathogenesis-associated pathways (such as mitogen-activated protein kinase and vascular endothelial growth factor signaling pathway). Collectively, the data from our study provides novel insight into the ligament injury-related miRNA dysregulation in patients with OA.
Collapse
Affiliation(s)
- Bin Li
- Department of Orthopaedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Lunhao Bai
- Department of Orthopaedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Peng Shen
- Department of Orthopaedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yue Sun
- Department of Orthopaedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Zhizuo Chen
- Department of Orthopaedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yu Wen
- Department of Histology and Embryology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110122, P.R. China
| |
Collapse
|
104
|
Nieminen HJ, Barreto G, Finnilä MA, García-Pérez A, Salmi A, Ranjan S, Eklund KK, Pritzker KPH, Saarakkala S, Hæggström E. Laser-ultrasonic delivery of agents into articular cartilage. Sci Rep 2017; 7:3991. [PMID: 28638116 PMCID: PMC5479804 DOI: 10.1038/s41598-017-04293-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 05/12/2017] [Indexed: 11/09/2022] Open
Abstract
Research is ongoing to develop drug therapies to manage osteoarthritis (OA) and articular cartilage (AC) injuries. However, means to deliver drug to localized AC lesions are highly limited and not clinically available. This study investigates the capability of laser ultrasound (laser-induced plasma sound source) to deliver agents (methylene blue, MB, in PBS) into bovine AC. Treatment samples (n = 10) were immersed in MB solution simultaneously with LU exposure, while adjacent control 1 tissue (n = 10) was pre-treated with LU followed by immersion in MB and adjacent control 2 tissue (n = 10) was only immersed in MB. AC exposed (n = 22) or not exposed (n = 27) to LU were characterized for anomalies in structure, composition, viability or RNA expression. Optically detected MB content was significantly (p < 0.01) higher in treatment samples up to a depth of 500 µm from AC surface as compared to controls. No major unwanted short-term effects on AC structure, proteoglycan or collagen contents, chondrocyte viability or RNA expression levels were detected. In conclusion, LU can deliver agents into AC without major short-term concerns on safety. LU could reveal new strategies for the development of localized drug therapies in AC.
Collapse
Affiliation(s)
- Heikki J Nieminen
- Electronics Research Laboratory, Department of Physics, University of Helsinki, Helsinki, Finland.
- Research Group of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
| | - Gonçalo Barreto
- Orton Orthopaedic Hospital and Research Institute, Invalid Foundation, Helsinki, Finland
- Department of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikko A Finnilä
- Research Group of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - Alejandro García-Pérez
- Electronics Research Laboratory, Department of Physics, University of Helsinki, Helsinki, Finland
- Department of Electronic Engineering, Higher Technological Institute of Poza Rica, Poza Rica, México, USA
| | - Ari Salmi
- Electronics Research Laboratory, Department of Physics, University of Helsinki, Helsinki, Finland
| | - Sanjeev Ranjan
- Laboratory of Radiochemistry, Department of Chemistry, University of Helsinki, Helsinki, Finland
| | - Kari K Eklund
- Department of Rheumatology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Kenneth P H Pritzker
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, Mount Sinai Hospital, Toronto, Canada
| | - Simo Saarakkala
- Research Group of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
| | - Edward Hæggström
- Electronics Research Laboratory, Department of Physics, University of Helsinki, Helsinki, Finland
| |
Collapse
|
105
|
The Role of Low-Intensity Pulsed Ultrasound on Cartilage Healing in Knee Osteoarthritis: A Review. PM R 2017; 9:1268-1277. [PMID: 28606838 DOI: 10.1016/j.pmrj.2017.05.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 04/05/2017] [Accepted: 05/25/2017] [Indexed: 10/19/2022]
Abstract
Ultrasound (US) is a therapeutic modality that has been used in the treatment of musculoskeletal conditions for decades. In recent years, there have been technological advancements using low-intensity pulsed ultrasound (LIPUS) as a clinical modality. The purpose of this review was to critically examine the medical literature to determine the effects of LIPUS on the chondrogenic properties of knee osteoarthritis. A literature search of 3 major databases (PubMed, Scopus, and EMBASE) was performed. Two independent physician reviewers screened titles and abstracts, yielding a total of 18 relevant articles after the inclusion and exclusion criteria were applied. Results favored that LIPUS has a promising effect on the cellular elements in articular cartilage, specifically on chondrocytes in knee osteoarthritis. Although the use of LIPUS is encouraging based on basic science and preclinical data, there is a paucity of evidence with respect to humans. Consequently, there is insufficient evidence to recommend for or against LIPUS in clinical OA populations. We suggest future directions for research centered on LIPUS in both human and animal models to delineate the effect on the biologic properties of cartilage in knee osteoarthritis. LEVEL OF EVIDENCE III.
Collapse
|
106
|
Statistically optimized fast dissolving microneedle transdermal patch of meloxicam: A patient friendly approach to manage arthritis. Eur J Pharm Sci 2017; 104:114-123. [DOI: 10.1016/j.ejps.2017.04.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/30/2017] [Accepted: 04/01/2017] [Indexed: 02/06/2023]
|
107
|
Nganvongpanit K, Soponteerakul R, Kaewkumpai P, Punyapornwithaya V, Buddhachat K, Nomsiri R, Kaewmong P, Kittiwatanawong K, Chawangwongsanukun R, Angkawanish T, Thitaram C, Mahakkanukrauh P. Osteoarthritis in two marine mammals and 22 land mammals: learning from skeletal remains. J Anat 2017; 231:140-155. [PMID: 28542897 DOI: 10.1111/joa.12620] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2017] [Indexed: 11/28/2022] Open
Abstract
The occurrence of osteoarthritis (OA) in marine mammals is still questionable. Here we investigated the prevalence of OA in marine (dolphin and dugong) and terrestrial mammals (Asian elephant, Asiatic buffalo, camel, cat, cattle, deer, dog, domestic goat, horse, human, hyena, impala, lion, Malayan tapir, Assam macaque, mule, pig, rabbit, red kangaroo, sheep, tiger and waterbuck). Skeletal remains obtained from five institutes were used as subjects; a total of 45 different parts (locations) of bones were observed for OA lesions. The prevalence of OA was reported as number of OA lesions/total number of bones. Our results revealed that the presence of OA in marine species (dolphin and dugong) was 2.44% and 3.33%, respectively. In dolphins, the highest OA occurrence was on the left and right humeral trochlea, with 13.68% and 12.63%, respectively, while the highest number of OA lesions in dugongs was on the lumbar vertebrae (8.79%). No significant difference (P > 0.05) in the prevalence of OA between sexes in dolphins and dugongs was observed, but we found a significant difference (P < 0.05) in 24 bone locations of human bones, which had the highest OA prevalence (48.93%), followed by dogs (3.94%). In conclusion, OA can occur in marine mammals, similar to terrestrial mammals, even though their natural habitat is the ocean.
Collapse
Affiliation(s)
- Korakot Nganvongpanit
- Animal Bone and Joint Research Laboratory, Department of Veterinary Biosciences and Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Ratsadakorn Soponteerakul
- Animal Bone and Joint Research Laboratory, Department of Veterinary Biosciences and Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Piyatida Kaewkumpai
- Animal Bone and Joint Research Laboratory, Department of Veterinary Biosciences and Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Veerasak Punyapornwithaya
- Department of Food Animal Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Kittisak Buddhachat
- Department of Biology, Faculty of Science, Naresuan University, Phitsanulok, Thailand
| | - Raksiri Nomsiri
- Veterinary Conservation and Research Section, Chiang Mai Night Safari, Chiang Mai, Thailand
| | | | | | - Rachanchai Chawangwongsanukun
- Department of Pre-clinical Sciences and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Taweepoke Angkawanish
- National Elephant Institute, Forest Industry Organization, Hang Chat, Lampang, Thailand
| | - Chatchote Thitaram
- Elephant Research and Education Center, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pasuk Mahakkanukrauh
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Excellence Center in Osteology Research and Training Center, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
108
|
Cartilage breakdown in microgravity-a problem for long-term spaceflight? NPJ Regen Med 2017; 2:10. [PMID: 29302346 PMCID: PMC5677769 DOI: 10.1038/s41536-017-0016-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 02/13/2017] [Accepted: 03/06/2017] [Indexed: 12/02/2022] Open
|
109
|
Tan AR, Hung CT. Concise Review: Mesenchymal Stem Cells for Functional Cartilage Tissue Engineering: Taking Cues from Chondrocyte-Based Constructs. Stem Cells Transl Med 2017; 6:1295-1303. [PMID: 28177194 PMCID: PMC5442836 DOI: 10.1002/sctm.16-0271] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 12/21/2016] [Indexed: 01/01/2023] Open
Abstract
Osteoarthritis, the most prevalent form of joint disease, afflicts 9% of the U.S. population over the age of 30 and costs the economy nearly $100 billion annually in healthcare and socioeconomic costs. It is characterized by joint pain and dysfunction, though the pathophysiology remains largely unknown. Due to its avascular nature and limited cellularity, articular cartilage exhibits a poor intrinsic healing response following injury. As such, significant research efforts are aimed at producing engineered cartilage as a cell-based approach for articular cartilage repair. However, the knee joint is mechanically demanding, and during injury, also a milieu of harsh inflammatory agents. The unforgiving mechano-chemical environment requires tissue replacements that are capable of bearing such burdens. The use of mesenchymal stem cells (MSCs) for cartilage tissue engineering has emerged as a promising cell source due to their ease of isolation, capacity to readily expand in culture, and ability to undergo lineage-specific differentiation into chondrocytes. However, to date, very few studies utilizing MSCs have successfully recapitulated the structural and functional properties of native cartilage, exposing the difficult process of uniformly differentiating stem cells into desired cell fates and maintaining the phenotype during in vitro culture and after in vivo implantation. To address these shortcomings, here, we present a concise review on modulating stem cell behavior, tissue development and function using well-developed techniques from chondrocyte-based cartilage tissue engineering. Stem Cells Translational Medicine 2017;6:1295-1303.
Collapse
|
110
|
Bonitsky CM, McGann ME, Selep MJ, Ovaert TC, Trippel SB, Wagner DR. Genipin crosslinking decreases the mechanical wear and biochemical degradation of impacted cartilage in vitro. J Orthop Res 2017; 35:558-565. [PMID: 27584857 PMCID: PMC5518482 DOI: 10.1002/jor.23411] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 08/29/2016] [Indexed: 02/04/2023]
Abstract
High energy trauma to cartilage causes surface fissures and microstructural damage, but the degree to which this damage renders the tissue more susceptible to wear and contributes to the progression of post-traumatic osteoarthritis (PTOA) is unknown. Additionally, no treatments are currently available to strengthen cartilage after joint trauma and to protect the tissue from subsequent degradation and wear. The purposes of this study were to investigate the role of mechanical damage in the degradation and wear of cartilage, to evaluate the effects of impact and subsequent genipin crosslinking on the changes in the viscoelastic parameters of articular cartilage, and to test the hypothesis that genipin crosslinking is an effective treatment to enhance the resistance to biochemical degradation and mechanical wear. Results demonstrate that cartilage stiffness decreases after impact loading, likely due to the formation of fissures and microarchitectural damage, and is partially or fully restored by crosslinking. The wear resistance of impacted articular cartilage was diminished compared to undamaged cartilage, suggesting that mechanical damage that is directly induced by the impact may contribute to the progression of PTOA. However, the decrease in wear resistance was completely reversed by the crosslinking treatments. Additionally, the crosslinking treatments improved the resistance to collagenase digestion at the impact-damaged articular surface. These results highlight the potential therapeutic value of collagen crosslinking via genipin in the prevention of cartilage degeneration after traumatic injury. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:558-565, 2017.
Collapse
Affiliation(s)
- Craig M. Bonitsky
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Megan E. McGann
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Michael J. Selep
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Timothy C. Ovaert
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Stephen B. Trippel
- Deparment of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Diane R. Wagner
- Department of Mechanical Engineering, Indiana University-Purdue University Indianapolis, 723 W. Michigan St. SL 260, Indianapolis, Indiana 46202
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana
| |
Collapse
|
111
|
Ramli ANM, Aznan TNT, Illias RM. Bromelain: from production to commercialisation. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2017; 97:1386-1395. [PMID: 27790704 DOI: 10.1002/jsfa.8122] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 09/26/2016] [Accepted: 10/25/2016] [Indexed: 06/06/2023]
Abstract
Bromelain is a mixture of proteolytic enzymes found in pineapple (Ananas comosus) plants. It can be found in several parts of the pineapple plant, including the stem, fruit, leaves and peel. High demand for bromelain has resulted in gradual increases in bromelain production. These increases have led to the need for a bromelain production strategy that yields more purified bromelain at a lower cost and with fewer production steps. Previously, bromelain was purified by conventional centrifugation, ultrafiltration and lyophilisation. Recently, the development of more modern purification techniques such as gel filtration, ion exchange chromatography, affinity chromatography, aqueous two-phase extraction and reverse micelle chromatography has resulted in increased industrial bromelain production worldwide. In addition, recombinant DNA technology has emerged as an alternative strategy for producing large amounts of ultrapure bromelain. An up-to-date compilation of data regarding the commercialisation of bromelain in the clinical, pharmaceutical and industrial fields is provided in this review. © 2016 Society of Chemical Industry.
Collapse
Affiliation(s)
- Aizi Nor Mazila Ramli
- Faculty of Industrial Science and Technology, Universiti Malaysia Pahang, Lebuhraya Tun Razak, 26300, Gambang, Kuantan, Pahang Darul Makmur, Malaysia
| | - Tuan Norsyalieza Tuan Aznan
- Faculty of Industrial Science and Technology, Universiti Malaysia Pahang, Lebuhraya Tun Razak, 26300, Gambang, Kuantan, Pahang Darul Makmur, Malaysia
| | - Rosli Md Illias
- Department of Bioprocess Engineering, Faculty of Chemical Engineering, Universiti Teknologi Malaysia, 81310, Skudai, Johor, Malaysia
| |
Collapse
|
112
|
O’Connell G, Garcia J, Amir J. 3D Bioprinting: New Directions in Articular Cartilage Tissue Engineering. ACS Biomater Sci Eng 2017; 3:2657-2668. [DOI: 10.1021/acsbiomaterials.6b00587] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Grace O’Connell
- Department
of Mechanical Engineering University of California, Berkeley, 5122 Etcheverry Hall, Berkeley, California 94720, United States
| | - Jeanette Garcia
- IBM Research-Almaden, 650
Harry Road K17/D2, San Jose, California 95120, United States
| | - Jamali Amir
- Joint Preservation Institute, 2825 J Street #440, Sacramento, California 95816, United States
| |
Collapse
|
113
|
Effectiveness of diclofenac versus paracetamol in knee osteoarthritis: a randomised controlled trial in primary care. Br J Gen Pract 2017. [PMID: 26212849 DOI: 10.3399/bjgp15x686101] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
BACKGROUND The effectiveness of diclofenac versus paracetamol in primary care patients with pain caused by knee osteoarthritis is unclear. AIM To assess the effectiveness of diclofenac compared with paracetamol over a period of 2, 4, and 12 weeks in patients with knee osteoarthritis. DESIGN AND SETTING Randomised controlled trial in general practice. METHOD There were 104 patients included in the study, they were aged ≥45 years consulting their GP with knee pain caused by knee osteoarthritis. Patients were randomly allocated to diclofenac (n = 52) or paracetamol (n = 52) for at least 2 weeks. Primary outcomes were daily knee pain severity, and knee pain and function measured with the Knee Injury and Osteoarthritis Outcome Score (KOOS). RESULTS Over a period of 2- and 4-weeks follow-up, no significant difference in daily knee pain was found between the patient groups: estimated differences of 0.5 (95% CI = -0.2 to 1.3) and -0.2 (95% CI = -1.0 to 0.7), respectively. Over the 12-weeks follow-up, no significant differences were found between both groups for KOOS pain: estimated difference of -2.8 (95% CI = -10.7 to 5.1) and KOOS function of -2.7 (-10.6 to 5.0). CONCLUSION Over a period of 2- and 4-weeks follow-up no significant difference in daily measured knee pain severity was found between primary care patients with knee osteoarthritis taking paracetamol or diclofenac. Also, over a period of 12-weeks follow-up no significant differences were found regarding KOOS pain and KOOS function between both groups. Patients more frequently reported minor adverse events after taking diclofenac (64%) than paracetamol (46%).
Collapse
|
114
|
Engelhardt C, Farron A, Becce F, Place N, Pioletti DP, Terrier A. Effects of glenoid inclination and acromion index on humeral head translation and glenoid articular cartilage strain. J Shoulder Elbow Surg 2017; 26:157-164. [PMID: 27522337 DOI: 10.1016/j.jse.2016.05.031] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 05/17/2016] [Accepted: 05/26/2016] [Indexed: 02/01/2023]
Abstract
BACKGROUND Previous clinical studies have reported associations between glenoid inclination (GI), the acromion index (AI), and the critical shoulder angle (CSA) on the one hand and the occurrence of glenohumeral osteoarthritis and supraspinatus tendon tears on the other hand. The objective of this work was to analyze the correlations and relative importance of these different anatomic parameters. METHODS Using a musculoskeletal shoulder model developed from magnetic resonance imaging scans of 1 healthy volunteer, we varied independently GI from 0° to 15° and AI from 0.5 to 0.8. The corresponding CSA varied from 20.9° to 44.1°. We then evaluated humeral head translation and critical strain volume in the glenoid articular cartilage at 60° of abduction in the scapular plane. These values were correlated with GI, AI, and CSA. RESULTS Humeral head translation was positively correlated with GI (R = 0.828, P < .0001), AI (R = 0.539, P < .0001), and CSA (R = 0.964, P < .0001). Glenoid articular cartilage strain was also positively correlated with GI (R = 0.489, P = .0004) but negatively with AI (R = -0.860, P < .0001) and CSA (R = -0.285, P < .0473). CONCLUSIONS The biomechanical shoulder model is consistent with clinical observations. The prediction strength of CSA is confirmed for humeral head translation and thus presumably for rotator cuff tendon tears, whereas the AI seems more appropriate to evaluate the risk of glenohumeral osteoarthritis caused by excessive articular cartilage strain. As a next step, we should corroborate these theoretical findings with clinical data.
Collapse
Affiliation(s)
- Christoph Engelhardt
- Laboratory of Biomechanical Orthopedics, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Alain Farron
- Service of Orthopedics and Traumatology, Lausanne University Hospital, Lausanne, Switzerland
| | - Fabio Becce
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital, Lausanne, Switzerland
| | - Nicolas Place
- Institute of Sports Sciences, Faculty of Biology Medicine, University of Lausanne, Lausanne, Switzerland; Department of Physiology, Faculty of Biology Medicine, University of Lausanne, Lausanne, Switzerland
| | - Dominique P Pioletti
- Laboratory of Biomechanical Orthopedics, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Alexandre Terrier
- Laboratory of Biomechanical Orthopedics, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| |
Collapse
|
115
|
Mumme M, Steinitz A, Nuss KM, Klein K, Feliciano S, Kronen P, Jakob M, von Rechenberg B, Martin I, Barbero A, Pelttari K. Regenerative Potential of Tissue-Engineered Nasal Chondrocytes in Goat Articular Cartilage Defects. Tissue Eng Part A 2016; 22:1286-1295. [DOI: 10.1089/ten.tea.2016.0159] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Marcus Mumme
- Department of Biomedicine, University Hospital of Basel, University of Basel, Basel, Switzerland
- Clinic for Traumatologic Surgery, University Hospital of Basel, Basel, Switzerland
| | - Amir Steinitz
- Department of Biomedicine, University Hospital of Basel, University of Basel, Basel, Switzerland
- Clinic for Traumatologic Surgery, University Hospital of Basel, Basel, Switzerland
| | - Katja M. Nuss
- Musculoskeletal Research Unit (MSRU), Equine Department, University of Zurich, Zürich, Switzerland
| | - Karina Klein
- Musculoskeletal Research Unit (MSRU), Equine Department, University of Zurich, Zürich, Switzerland
| | - Sandra Feliciano
- Department of Biomedicine, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Peter Kronen
- Competence Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Zürich, Switzerland
- Veterinary Anaesthesia Services–International (VAS), Winterthur, Switzerland
| | - Marcel Jakob
- Clinic for Traumatologic Surgery, University Hospital of Basel, Basel, Switzerland
| | - Brigitte von Rechenberg
- Musculoskeletal Research Unit (MSRU), Equine Department, University of Zurich, Zürich, Switzerland
- Competence Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Zürich, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Andrea Barbero
- Department of Biomedicine, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Karoliina Pelttari
- Department of Biomedicine, University Hospital of Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
116
|
Mumme M, Barbero A, Miot S, Wixmerten A, Feliciano S, Wolf F, Asnaghi AM, Baumhoer D, Bieri O, Kretzschmar M, Pagenstert G, Haug M, Schaefer DJ, Martin I, Jakob M. Nasal chondrocyte-based engineered autologous cartilage tissue for repair of articular cartilage defects: an observational first-in-human trial. Lancet 2016; 388:1985-1994. [PMID: 27789021 DOI: 10.1016/s0140-6736(16)31658-0] [Citation(s) in RCA: 177] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 08/24/2016] [Accepted: 08/25/2016] [Indexed: 10/20/2022]
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.
Collapse
Affiliation(s)
- Marcus Mumme
- Department of Surgery and Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Andrea Barbero
- Department of Surgery and Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sylvie Miot
- Department of Surgery and Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Anke Wixmerten
- Department of Surgery and Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sandra Feliciano
- Department of Surgery and Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Francine Wolf
- Department of Surgery and Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Adelaide M Asnaghi
- Department of Surgery and Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Daniel Baumhoer
- Department of Institute of Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Oliver Bieri
- Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Martin Kretzschmar
- Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Geert Pagenstert
- Department of Surgery and Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Martin Haug
- Department of Surgery and Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Dirk J Schaefer
- Department of Surgery and Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Ivan Martin
- Department of Surgery and Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Marcel Jakob
- Department of Surgery and Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
117
|
Nowicki MA, Castro NJ, Plesniak MW, Zhang LG. 3D printing of novel osteochondral scaffolds with graded microstructure. NANOTECHNOLOGY 2016; 27:414001. [PMID: 27606933 DOI: 10.1088/0957-4484/27/41/414001] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Osteochondral tissue has a complex graded structure where biological, physiological, and mechanical properties vary significantly over the full thickness spanning from the subchondral bone region beneath the joint surface to the hyaline cartilage region at the joint surface. This presents a significant challenge for tissue-engineered structures addressing osteochondral defects. Fused deposition modeling (FDM) 3D bioprinters present a unique solution to this problem. The objective of this study is to use FDM-based 3D bioprinting and nanocrystalline hydroxyapatite for improved bone marrow human mesenchymal stem cell (hMSC) adhesion, growth, and osteochondral differentiation. FDM printing parameters can be tuned through computer aided design and computer numerical control software to manipulate scaffold geometries in ways that are beneficial to mechanical performance without hindering cellular behavior. Additionally, the ability to fine-tune 3D printed scaffolds increases further through our investment casting procedure which facilitates the inclusion of nanoparticles with biochemical factors to further elicit desired hMSC differentiation. For this study, FDM was used to print investment-casting molds innovatively designed with varied pore distribution over the full thickness of the scaffold. The mechanical and biological impacts of the varied pore distributions were compared and evaluated to determine the benefits of this physical manipulation. The results indicate that both mechanical properties and cell performance improve in the graded pore structures when compared to homogeneously distributed porous and non-porous structures. Differentiation results indicated successful osteogenic and chondrogenic manipulation in engineered scaffolds.
Collapse
Affiliation(s)
- Margaret A Nowicki
- Department of Mechanical and Aerospace Engineering, The George Washington University, 800 22nd Street, NW, Washington, DC, 20052, USA
| | | | | | | |
Collapse
|
118
|
Olive and grape seed extract prevents post-traumatic osteoarthritis damages and exhibits in vitro anti IL-1β activities before and after oral consumption. Sci Rep 2016; 6:33527. [PMID: 27640363 PMCID: PMC5027597 DOI: 10.1038/srep33527] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/26/2016] [Indexed: 12/24/2022] Open
Abstract
Polyphenols exert a large range of beneficial effects in the prevention of age-related diseases. We sought to determine whether an extract of olive and grape seed standardized according to hydroxytyrosol (HT) and procyanidins (PCy) content, exerts preventive anti-osteoathritic effects. To this aim, we evaluated whether the HT/PCy mix could (i) have in vitro anti-inflammatory and chondroprotective actions, (ii) exert anti-osteoarthritis effects in two post-traumatic animal models and (iii) retain its bioactivity after oral administration. Anti-inflammatory and chondroprotective actions of HT/PCy were tested on primary cultured rabbit chondrocytes stimulated by interleukin-1 beta (IL-1β). The results showed that HT/PCy exerts anti-inflammatory and chondroprotective actions in vitro. The preventive effect of HT/PCy association was assessed in two animal models of post-traumatic OA in mice and rabbits. Diet supplementation with HT/PCy significantly decreased the severity of post-traumatic osteoarthritis in two complementary mice and rabbit models. The bioavailability and bioactivity was evaluated following gavage with HT/PCy in rabbits. Regular metabolites from HT/PCy extract were found in sera from rabbits following oral intake. Finally, sera from rabbits force-fed with HT/PCy conserved anti-IL-1β effect, suggesting the bioactivity of this extract. To conclude, HT/PCy extract may be of clinical significance for the preventive treatment of osteoarthritis.
Collapse
|
119
|
Nara H, Sato K, Kaieda A, Oki H, Kuno H, Santou T, Kanzaki N, Terauchi J, Uchikawa O, Kori M. Design, synthesis, and biological activity of novel, potent, and highly selective fused pyrimidine-2-carboxamide-4-one-based matrix metalloproteinase (MMP)-13 zinc-binding inhibitors. Bioorg Med Chem 2016; 24:6149-6165. [PMID: 27825552 DOI: 10.1016/j.bmc.2016.09.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 09/03/2016] [Accepted: 09/06/2016] [Indexed: 02/07/2023]
Abstract
Matrix metalloproteinase-13 (MMP-13), a member of the collagenase family of enzymes, has been implicated to play a key role in the pathology of osteoarthritis. Recently, we have reported the discovery of a series of quinazoline-2-carboxamide based non-zinc-binding MMP-13 selective inhibitors, as exemplified by compound 1. We then continued our research of a novel class of zinc-binding inhibitors to obtain follow-up compounds with different physicochemical, pharmacokinetic, and biological activity profiles. In order to design selective MMP-13 inhibitors, we adopted a strategy of connecting a zinc-binding group with the quinazoline-2-carboxamide system, a unique S1' binder, by an appropriate linker. Among synthesized compounds, a triazolone inhibitor 35 exhibited excellent potency (IC50=0.071nM) and selectivity (greater than 170-fold) over other MMPs (MMP-1, 2, 3, 7, 8, 9, 10, 12, and 14) and tumor necrosis factor-α converting enzyme (TACE). In this article, the design, synthesis, and biological activity of novel zinc-binding MMP-13 inhibitors are described.
Collapse
Affiliation(s)
- Hiroshi Nara
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Kenjiro Sato
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Akira Kaieda
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Hideyuki Oki
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Haruhiko Kuno
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Takashi Santou
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Naoyuki Kanzaki
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Jun Terauchi
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Osamu Uchikawa
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Masakuni Kori
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| |
Collapse
|
120
|
Parathyroid hormone 1-34 reduces dexamethasone-induced terminal differentiation in human articular chondrocytes. Toxicology 2016; 368-369:116-128. [PMID: 27608943 DOI: 10.1016/j.tox.2016.09.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 09/04/2016] [Indexed: 11/24/2022]
Abstract
Intra-articular injection of dexamethasone (Dex) is occasionally used to relieve pain and inflammation in osteoarthritis (OA) patients. Dex induces terminal differentiation of chondrogenic mesenchymal stem cells in vitro and causes impaired longitudinal skeletal growth in vivo. Parathyroid hormone 1-34 (PTH 1-34) has been shown to reverse terminal differentiation of osteoarthritic articular chondrocytes. We hypothesized that Dex induces terminal differentiation of articular chondrocytes and that this effect can be mitigated by PTH 1-34 treatment. We tested the effect of Dex on terminal differentiation in human articular chondrocytes and further tested if PTH 1-34 reverses the effects. We found that Dex treatment downregulated chondrogenic-induced expressions of SOX-9, collagen type IIa1 (Col2a1), and aggrecan and reduced synthesis of cartilaginous matrix (Col2a1 and sulfated glycosaminoglycan) synthesis. Dex treatment upregulated chondrocyte hypertrophic markers of collagen type X and alkaline phosphatase at mRNA and protein levels, and it increased the cell size of articular chondrocytes and induced cell death. These results indicated that Dex induces terminal differentiation of articular chondrocytes. To test whether PTH 1-34 treatment reverses Dex-induced terminal differentiation of articular chondrocytes, PTH 1-34 was co-administered with Dex. Results showed that PTH 1-34 treatment reversed both changes of chondrogenic and hypertrophic markers in chondrocytes induced by Dex. PTH 1-34 also decreased Dex-induced cell death. PTH 1-34 treatment reduces Dex-induced terminal differentiation and apoptosis of articular chondrocytes, and PTH 1-34 treatment may protect articular cartilage from further damage when received Dex administration.
Collapse
|
121
|
Rautiainen J, Nieminen MT, Salo EN, Kokkonen HT, Mangia S, Michaeli S, Gröhn O, Jurvelin JS, Töyräs J, Nissi MJ. Effect of collagen cross-linking on quantitative MRI parameters of articular cartilage. Osteoarthritis Cartilage 2016; 24:1656-64. [PMID: 27143363 DOI: 10.1016/j.joca.2016.04.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 03/31/2016] [Accepted: 04/23/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To investigate the sensitivity of quantitative magnetic resonance imaging (MRI) parameters to increase of collagen cross-linking in articular cartilage, a factor possibly contributing to the aging-related development of osteoarthritis (OA). The issue has not been widely studied although collagen cross-links may significantly affect the evaluation of cartilage imaging outcome. DESIGN Osteochondral samples (n = 14) were prepared from seven bovine patellae. To induce cross-linking, seven samples were incubated in threose while the other seven served as non-treated controls. The specimens were scanned at 9.4 T for T1, T1Gd (dGEMRIC), T2, adiabatic and continuous wave (CW) T1ρ, adiabatic T2ρ and T1sat relaxation times. Specimens from adjacent tissue were identically treated and used for reference to determine biomechanical properties, collagen, proteoglycan and cross-link contents, fixed charge density (FCD), collagen fibril anisotropy and water concentration of cartilage. RESULTS In the threose-treated sample group, cross-links (pentosidine, lysyl pyridinoline (LP)), FCD and equilibrium modulus were significantly (P < 0.05) higher as compared to the non-treated group. Threose treatment resulted in significantly greater T1Gd relaxation time constant (+26%, P < 0.05), although proteoglycan content was not altered. Adiabatic and CW-T1ρ were also significantly increased (+16%, +28%, P < 0.05) while pre-contrast T1 was significantly decreased (-10%, P < 0.05) in the threose group. T2, T2ρ and T1sat did not change significantly. CONCLUSION Threose treatment induced collagen cross-linking and changes in the properties of articular cartilage, which were detected by T1, T1Gd and T1ρ relaxation time constants. Cross-linking should be considered especially when interpreting the outcome of contrast-enhanced MRI in aging populations.
Collapse
Affiliation(s)
- J Rautiainen
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland; Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland; Medical Research Center Oulu, University of Oulu, Oulu, Finland.
| | - M T Nieminen
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland; Medical Research Center Oulu, University of Oulu, Oulu, Finland; Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland.
| | - E-N Salo
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland.
| | - H T Kokkonen
- Diagnostic Imaging Center, Kuopio University Hospital, Kuopio, Finland.
| | - S Mangia
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA.
| | - S Michaeli
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA.
| | - O Gröhn
- Department of Neurobiology, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| | - J S Jurvelin
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland; Diagnostic Imaging Center, Kuopio University Hospital, Kuopio, Finland.
| | - J Töyräs
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland; Diagnostic Imaging Center, Kuopio University Hospital, Kuopio, Finland.
| | - M J Nissi
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland; Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland; Medical Research Center Oulu, University of Oulu, Oulu, Finland; Diagnostic Imaging Center, Kuopio University Hospital, Kuopio, Finland.
| |
Collapse
|
122
|
Olsen AL, Strand LI, Skjaerven LH, Sundal MA, Magnussen LH. Patient education and basic body awareness therapy in hip osteoarthritis – a qualitative study of patients’ movement learning experiences. Disabil Rehabil 2016; 39:1631-1638. [DOI: 10.1080/09638288.2016.1209578] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Aarid Liland Olsen
- Department of Physiotherapy, Haukeland University Hospital, Bergen, Norway
| | - Liv Inger Strand
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Liv Helvik Skjaerven
- Department of Health and Social Sciences, Bergen University College, Bergen, Norway
| | - Mary-Anne Sundal
- Department of Health and Social Sciences, Bergen University College, Bergen, Norway
| | - Liv Heide Magnussen
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
- Department of Health and Social Sciences, Bergen University College, Bergen, Norway
| |
Collapse
|
123
|
Usmani SE, Ulici V, Pest MA, Hill TL, Welch ID, Beier F. Context-specific protection of TGFα null mice from osteoarthritis. Sci Rep 2016; 6:30434. [PMID: 27457421 PMCID: PMC4960644 DOI: 10.1038/srep30434] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 07/05/2016] [Indexed: 01/30/2023] Open
Abstract
Transforming growth factor alpha (TGFα) is a growth factor involved in osteoarthritis (OA). TGFα induces an OA-like phenotype in articular chondrocytes, by inhibiting matrix synthesis and promoting catabolic factor expression. To better understand TGFα’s potential as a therapeutic target, we employed two in vivo OA models: (1) post-traumatic and (2) aging related OA. Ten-week old and six-month old male Tgfa null mice and their heterozygous (control) littermates underwent destabilization of the medial meniscus (DMM) surgery. Disease progression was assessed histologically using the Osteoarthritis Research Society International (OARSI) scoring system. As well, spontaneous disease progression was analyzed in eighteen-month-old Tgfa null and heterozygous mice. Ten-week old Tgfa null mice were protected from OA progression at both seven and fourteen weeks post-surgery. No protection was seen however in six-month old null mice after DMM surgery, and no differences were observed between genotypes in the aging model. Thus, young Tgfa null mice are protected from OA progression in the DMM model, while older mice are not. In addition, Tgfa null mice are equally susceptible to spontaneous OA development during aging. Thus, TGFα might be a valuable therapeutic target in some post-traumatic forms of OA, however its role in idiopathic disease is less clear.
Collapse
Affiliation(s)
- Shirine E Usmani
- Department of Physiology &Pharmacology, Schulich School of Medicine &Dentistry, The University of Western Ontario, London, ON, Canada
| | - Veronica Ulici
- Department of Physiology &Pharmacology, Schulich School of Medicine &Dentistry, The University of Western Ontario, London, ON, Canada
| | - Michael A Pest
- Department of Physiology &Pharmacology, Schulich School of Medicine &Dentistry, The University of Western Ontario, London, ON, Canada
| | - Tracy L Hill
- Department of Animal Care and Veterinary Services, University of Western Ontario, London, Canada
| | - Ian D Welch
- Department of Animal Care and Veterinary Services, University of Western Ontario, London, Canada
| | - Frank Beier
- Department of Physiology &Pharmacology, Schulich School of Medicine &Dentistry, The University of Western Ontario, London, ON, Canada
| |
Collapse
|
124
|
Oinas J, Rieppo L, Finnilä MAJ, Valkealahti M, Lehenkari P, Saarakkala S. Imaging of Osteoarthritic Human Articular Cartilage using Fourier Transform Infrared Microspectroscopy Combined with Multivariate and Univariate Analysis. Sci Rep 2016; 6:30008. [PMID: 27445254 PMCID: PMC4956759 DOI: 10.1038/srep30008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 06/29/2016] [Indexed: 12/16/2022] Open
Abstract
The changes in chemical composition of human articular cartilage (AC) caused by osteoarthritis (OA) were investigated using Fourier transform infrared microspectroscopy (FTIR-MS). We demonstrate the sensitivity of FTIR-MS for monitoring compositional changes that occur with OA progression. Twenty-eight AC samples from tibial plateaus were imaged with FTIR-MS. Hyperspectral images of all samples were combined for K-means clustering. Partial least squares regression (PLSR) analysis was used to compare the spectra with the OARSI grade (histopathological grading of OA). Furthermore, the amide I and the carbohydrate regions were used to estimate collagen and proteoglycan contents, respectively. Spectral peak at 1338 cm(-1) was used to estimate the integrity of the collagen network. The layered structure of AC was revealed using the carbohydrate region for clustering. Statistically significant correlation was observed between the OARSI grade and the collagen integrity in the superficial (r = -0.55) and the deep (r = -0.41) zones. Furthermore, PLSR models predicted the OARSI grade from the superficial (r = 0.94) and the deep (r = 0.77) regions of the AC with high accuracy. Obtained results suggest that quantitative and qualitative changes occur in the AC composition during OA progression, and these can be monitored by the use of FTIR-MS.
Collapse
Affiliation(s)
- J Oinas
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Finland.,Medical Research Center, University of Oulu and Oulu University Hospital, Finland
| | - L Rieppo
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Finland.,Medical Research Center, University of Oulu and Oulu University Hospital, Finland
| | - M A J Finnilä
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Finland.,Medical Research Center, University of Oulu and Oulu University Hospital, Finland.,Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - M Valkealahti
- Medical Research Center, University of Oulu and Oulu University Hospital, Finland.,Department of Surgery, Oulu University Hospital, Finland
| | - P Lehenkari
- Medical Research Center, University of Oulu and Oulu University Hospital, Finland.,Department of Surgery, Oulu University Hospital, Finland.,Research Group of Cancer and Translational Medicine, Faculty of Medicine, University of Oulu, Finland
| | - S Saarakkala
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Finland.,Medical Research Center, University of Oulu and Oulu University Hospital, Finland.,Department of Diagnostic Radiology, Oulu University Hospital, Finland
| |
Collapse
|
125
|
Cheng A, Gustafson AR, Schaner Tooley CE, Zhang M. BMP-9 dependent pathways required for the chondrogenic differentiation of pluripotent stem cells. Differentiation 2016; 92:298-305. [PMID: 27056281 DOI: 10.1016/j.diff.2016.03.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 03/17/2016] [Accepted: 03/29/2016] [Indexed: 01/20/2023]
Abstract
Current cartilage repair therapies focus on the delivery of chondrocytes differentiated from mesenchymal stem cells, and thus understanding the factors that promote chondrogenesis may lead to improved therapies. Several bone morphogenetic proteins (BMPs) have been implicated in chondrogenic differentiation and/or chondrocyte function. Although the signaling pathways downstream of BMPs have been studied in other systems, their role in chondrogenesis is less well characterized. Here, we investigated the effects of BMP-9 in chondroprogenitor cells. Compared to BMP-2 and BMP-6, we showed that BMP-9 was significantly more potent in inducing chondrogenic differentiation in mouse C3H10T1/2 and ATDC5 cells. Moreover, we demonstrated that BMP-9 induces the phosphorylation of SMAD1/5 in a dose and time dependent manner. Confocal immunofluorescence microscopy further demonstrated an accumulation of phosphorylated SMAD1/5 in the nuclei of BMP-9 treated cells. Consistent with activation of the SMAD signaling pathway, we also observed an up-regulation of Id1 and PAI-I expression. Importantly, we demonstrated that the simultaneous knockdown of SMAD1 and SMAD5 was able to inhibit chondrogenesis. Additionally, we also observed activation of p38 by BMP-9, and pharmacological inhibition of this pathway blocked chondrogenesis. In contrast, inhibition of p44/42 ERK had no effect. Finally, we tested the ability of Noggin to block the actions of BMP-9. While Noggin potently inhibited the ability of BMP-2 to mediate differentiation, it had no significant effect on BMP-9. Our findings provide a clearer understanding of the cellular pathways utilized by BMP-9 for chondrogenesis that may help improve current therapies for regenerative cartilage repair.
Collapse
Affiliation(s)
- Alan Cheng
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA.
| | - Amber Renee Gustafson
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA
| | | | - Mei Zhang
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA.
| |
Collapse
|
126
|
Meireles S, De Groote F, Reeves ND, Verschueren S, Maganaris C, Luyten F, Jonkers I. Knee contact forces are not altered in early knee osteoarthritis. Gait Posture 2016; 45:115-20. [PMID: 26979892 DOI: 10.1016/j.gaitpost.2016.01.016] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 12/11/2015] [Accepted: 01/17/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE This study calculated knee contact forces (KCF) and its relations with knee external knee adduction moments (KAM) and/or flexion moments (KFM) during the stance phase of gait in patients with early osteoarthritis (OA), classified based on early joint degeneration on Magnetic Resonance Imaging (MRI). We aimed at assessing if altered KCF are already present in early structural degeneration. DESIGN Three-dimensional motion and ground reaction force data in 59 subjects with medial compartment knee OA (N=23 established OA, N=16 early OA, N=20 controls) were used as input for a musculoskeletal model. KAM and KFM, and KCF were estimated using OpenSim software. RESULTS No significant differences were found between controls and subjects with early OA. In early OA patients, KAM significantly explained 69% of the variance associated with the first peaks KCF but only KFM contributed to the second peaks KCF. The multiple correlation, combining KAM and KFM, showed to be higher. However, only 20% of the variance of second peak KCF was explained by both moments in established OA. CONCLUSION KCF are not increased in patients with early OA, suggesting that knee joint overload is more a consequence of further joint degeneration in more advanced stages of OA. Additionally, our results clearly show that KAM is not sufficient to predict joint loading at the end of the stance, where KFM contributes substantially to the loading, especially in early OA.
Collapse
Affiliation(s)
- S Meireles
- Department of Kinesiology, KU Leuven, Belgium.
| | - F De Groote
- Department of Mechanical Engineering, KU Leuven, Belgium.
| | - N D Reeves
- School of Healthcare Science, Manchester Metropolitan University, United Kingdom.
| | - S Verschueren
- Department of Rehabilitation Sciences, KU Leuven, Belgium.
| | - C Maganaris
- School of Sport and Exercise Sciences, Liverpool John Moores University, United Kingdom.
| | - F Luyten
- Department of Development and Regeneration, KU Leuven, Belgium.
| | - I Jonkers
- Department of Kinesiology, KU Leuven, Belgium.
| |
Collapse
|
127
|
Fodor PB, Paulseth SG. Adipose Derived Stromal Cell (ADSC) Injections for Pain Management of Osteoarthritis in the Human Knee Joint. Aesthet Surg J 2016; 36:229-36. [PMID: 26238455 DOI: 10.1093/asj/sjv135] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND This safety and feasibility study used autologous adipose-derived stromal vascular cells (the stromal vascular fraction [SVF] of adipose tissue), to treat 8 osteoarthritic knees in 6 patients of grade I to III (K-L scale) with initial pain of 4 or greater on a 10-point Visual Analog Scale (VAS). OBJECTIVES The primary objective of the study was evaluation of the safety of intra-articular injection of SVF. The secondary objective was to assess initial feasibility for reduction of pain in osteoarthritic knees. METHODS Adipose-derived SVF cells were obtained through enzymatic disaggregation of lipoaspirate, resuspension in 3 mL of Lactated Ringer's Solution, and injection directly into the intra-articular space of the knee, with a mean of 14.1 million viable, nucleated SVF cells per knee. Metrics included monitoring of adverse events and preoperative to postoperative changes in the Western Ontario and McMaster Universities Arthritis Index (WOMAC), the VAS pain scale, range of motion (ROM), timed up-and-go (TUG), and MRI. RESULTS No infections, acute pain flares, or other adverse events were reported. At 3-months postoperative, there was a statistically significant improvement in WOMAC and VAS scores (P < .02 and P < .001, respectively), which was maintained at 1 year. Physical therapy measurements for ROM and TUG both improved from preoperative to 3-months postoperative. Standard MRI assessment from preoperative to 3-months postoperative showed no detectable structural differences. All patients attained full activity with decreased knee pain. CONCLUSIONS Autologous SVF was shown to be safe and to present a new potential therapy for reduction of pain for osteoarthritis of the knee. LEVEL OF EVIDENCE 4: Therapeutic.
Collapse
Affiliation(s)
- Peter B Fodor
- Dr Fodor is an Associate Clinical Professor of Plastic Surgery, UCLA Medical Center, Los Angeles, California. Dr Paulseth is an Adjunct Instructor of Clinical Physical Therapy, Division of Biokinesiology and Physical Therapy, University of Southern California, Los Angeles, California
| | - Stephen G Paulseth
- Dr Fodor is an Associate Clinical Professor of Plastic Surgery, UCLA Medical Center, Los Angeles, California. Dr Paulseth is an Adjunct Instructor of Clinical Physical Therapy, Division of Biokinesiology and Physical Therapy, University of Southern California, Los Angeles, California
| |
Collapse
|
128
|
Mickiewicz B, Shin SY, Pozzi A, Vogel HJ, Clark AL. Serum Metabolite Profiles Are Altered by Erlotinib Treatment and the Integrin α1-Null Genotype but Not by Post-Traumatic Osteoarthritis. J Proteome Res 2016; 15:815-25. [PMID: 26784366 DOI: 10.1021/acs.jproteome.5b00719] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The risk of developing post-traumatic osteoarthritis (PTOA) following joint injury is high. Furthering our understanding of the molecular mechanisms underlying PTOA and/or identifying novel biomarkers for early detection may help to improve treatment outcomes. Increased expression of integrin α1β1 and inhibition of epidermal growth factor receptor (EGFR) signaling protect the knee from spontaneous OA; however, the impact of the integrin α1β1/EGFR axis on PTOA is currently unknown. We sought to determine metabolic changes in serum samples collected from wild-type and integrin α1-null mice that underwent surgery to destabilize the medial meniscus and were treated with the EGFR inhibitor erlotinib. Following (1)H nuclear magnetic resonance spectroscopy, we generated multivariate statistical models that distinguished between the metabolic profiles of erlotinib- versus vehicle-treated mice and the integrin α1-null versus wild-type mouse genotype. Our results show the sex-dependent effects of erlotinib treatment and highlight glutamine as a metabolite that counteracts this treatment. Furthermore, we identified a set of metabolites associated with increased reactive oxygen species production, susceptibility to OA, and regulation of TRP channels in α1-null mice. Our study indicates that systemic pharmacological and genetic factors have a greater effect on serum metabolic profiles than site-specific factors such as surgery.
Collapse
Affiliation(s)
| | | | - Ambra Pozzi
- Department of Medicine, Vanderbilt University , Nashville, Tennessee 37232, United States.,Department of Medicine, Veterans Affairs Hospital , Nashville, Tennessee 37232, United States
| | | | - Andrea L Clark
- Department of Surgery, Cumming School of Medicine, University of Calgary , Calgary T2N 4N1, AB, Canada
| |
Collapse
|
129
|
Paul J, Barg A, Kretzschmar M, Pagenstert G, Studler U, Hügle T, Wegner NJ, Valderrabano V, Geurts J. Increased Osseous (99m)Tc-DPD Uptake in End-Stage Ankle Osteoarthritis: Correlation Between SPECT-CT Imaging and Histologic Findings. Foot Ankle Int 2015; 36:1438-47. [PMID: 26231199 DOI: 10.1177/1071100715596745] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND We analyzed the histopathologic findings in end-stage osteoarthritic ankle joint tissue that display increased uptake of bone-seeking radiotracer in single-photon emission computed tomography-computed tomography (SPECT-CT) imaging. METHODS Six consecutive patients with end-stage osteoarthritis undergoing total ankle replacement received preoperative SPECT-CT imaging using (99m)Technetium dicarboxypropane diphosphonate ((99m)Tc-DPD). Using imaging data for stratification, osteochondral tissue sections were prepared from SPECT-positive (+) and -negative (-) areas of tibial and talar resection specimens. Histomorphometric analyses of osteoblast numbers, collagen deposition, and cartilage degeneration were performed on hematoxylin and eosin, van Gieson's and Safranin-O stained tissue sections. Osteoclast activity was visualized using tartrate-resistant acid phosphatase (TRAP) staining. RESULTS Increased (99m)Tc-DPD uptake was observed exclusively subjacent to the subchondral bone plate of tibial and talar joint compartments. SPECT(-) tissues displayed typical fatty marrow morphology containing mainly collagen-positive blood vessels and few marrow and bone-lining cells. SPECT(+) tissues were characterized by increased numbers of active bone-lining osteoblasts depositing collagen fibers. Collagen area fraction of subchondral bone marrow was significantly increased in SPECT(+) (0.52 ± 0.21) compared with SPECT(-) (0.29 ± 0.13) tissues (P = .30). Multinucleated TRAP(+) osteoclasts were absent from bone formation sites, but associated with vascular structures invading articular cartilage through the subchondral bone plate. Increased (99m)Tc-DPD uptake was specifically and strongly correlated with increased osteoblast numbers (P = .011), and with collagen area fraction (P = .030) but not with Mankin score (P = .202), or with osteoclast number (P = .576). CONCLUSION Subchondral bone tissues in SPECT(+) areas of end-stage ankle osteoarthritis were histologically characterized by increased osteoblast-mediated bone formation in the absence of functional osteoclasts, and increased cellularity and collagen deposition in marrow tissues. CLINICAL SIGNIFICANCE Our findings suggest a pathologic bone-remodeling process in end-stage ankle OA areas with increased (99m)Tc-DPD uptake.
Collapse
Affiliation(s)
- Jochen Paul
- Praxisklinik Rennbahn AG, Basel, Switzerland
| | | | | | | | - Ueli Studler
- University Hospital of Basel, Basel, Switzerland
| | - Thomas Hügle
- University Hospital of Basel, Basel, Switzerland
| | | | | | | |
Collapse
|
130
|
Pelttari K, Barbero A, Martin I. A potential role of homeobox transcription factors in osteoarthritis. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:254. [PMID: 26605300 DOI: 10.3978/j.issn.2305-5839.2015.09.44] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
When a healthy joint progressively becomes osteoarthritic, the structures of the affected cartilage, bone and synovia undergo an initial phase of rearrangement. The exact molecular and cellular events occurring in this early osteoarthritic transition phase still remain elusive. Homeobox (Hox) genes encode for transcription factors that typically regulate limb morphogenesis and skeletal formation during development. More recently they were shown to be required for tissue remodelling and homeostasis in adults and to be modulated in a variety of pathologies. Here we present and discuss the hypothesis that dysregulation of specific Hox genes is associated with the onset and development of osteoarthritis (OA). Discovering mechanisms modulating Hox gene expression could not only provide important information in understanding OA pathology and its initiation, but also help to identify biomarkers reflecting the state of early OA. This knowledge would allow anticipating the time window for clinical treatment of the affected cartilage and assist in the development of innovative strategies to restore joint homeostasis, e.g., by cell or gene therapy.
Collapse
Affiliation(s)
- Karoliina Pelttari
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Andrea Barbero
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel and University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| |
Collapse
|
131
|
Wyles CC, Houdek MT, Behfar A, Sierra RJ. Mesenchymal stem cell therapy for osteoarthritis: current perspectives. Stem Cells Cloning 2015; 8:117-24. [PMID: 26357483 PMCID: PMC4559256 DOI: 10.2147/sccaa.s68073] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Osteoarthritis (OA) is a painful chronic condition with a significant impact on quality of life. The societal burden imposed by OA is increasing in parallel with the aging population; however, no therapies have demonstrated efficacy in preventing the progression of this degenerative joint disease. Current mainstays of therapy include activity modification, conservative pain management strategies, weight loss, and if necessary, replacement of the affected joint. Mesenchymal stem cells (MSCs) are a multipotent endogenous population of progenitors capable of differentiation to musculoskeletal tissues. MSCs have a well-documented immunomodulatory role, managing the inflammatory response primarily through paracrine signaling. Given these properties, MSCs have been proposed as a potential regenerative cell therapy source for patients with OA. Research efforts are focused on determining the ideal source for derivation, as MSCs are native to several tissues. Furthermore, optimizing the mode of delivery remains a challenge both for appropriate localization of MSCs and for directed guidance toward stemming the local inflammatory process and initiating a regenerative response. Scaffolds and matrices with growth factor adjuvants may prove critical in this effort. The purpose of this review is to summarize the current state of MSC-based therapeutics for OA and discuss potential barriers that must be overcome for successful implementation of cell-based therapy as a routine treatment strategy in orthopedics.
Collapse
Affiliation(s)
- Cody C Wyles
- Mayo Medical School, Mayo Clinic, Rochester, MN, USA
| | - Matthew T Houdek
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Atta Behfar
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Rafael J Sierra
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
132
|
Runhaar J, van Middelkoop M, Reijman M, Willemsen S, Oei EH, Vroegindeweij D, van Osch G, Koes B, Bierma-Zeinstra SMA. Prevention of knee osteoarthritis in overweight females: the first preventive randomized controlled trial in osteoarthritis. Am J Med 2015; 128:888-895.e4. [PMID: 25818496 DOI: 10.1016/j.amjmed.2015.03.006] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 03/11/2015] [Accepted: 03/11/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND With accumulating knowledge on osteoarthritis development, the next step is to focus on possibilities for primary prevention. METHODS In a 2 × 2 factorial design, the effects of a diet-and-exercise program and of oral glucosamine sulfate (double blind and placebo-controlled) on the incidence of knee osteoarthritis were evaluated in a high-risk group of 407 middle-aged women with a body mass index ≥ 27 kg/m(2) without clinical signs of knee osteoarthritis at baseline (ISRCTN 42823086). Primary outcome was the incidence of knee osteoarthritis, defined as Kellgren & Lawrence grade ≥ 2, joint space narrowing of ≥ 1.0 mm, or clinical knee osteoarthritis (clinical and radiographic American College of Rheumatology criteria) after 2.5 years. RESULTS After 2.5 years, only 10% of all subjects were lost to follow-up, and 17% of all knees showed incident knee osteoarthritis. Accounting for the significant interaction between the interventions, no significant main effect of either intervention was found. Independently, both interventions alone showed indications of reduced knee osteoarthritis incidence (odds ratio [OR] 0.69; 95% CI, 0.39-1.21 for the diet-and-exercise program and OR 0.60; 95% CI, 0.31-1.12 for the glucosamine intervention). These effects were neutralized in subjects receiving both interventions (OR 0.97; 95% CI, 0.55-1.71). CONCLUSIONS No significant main effects of the diet-and-exercise program and of glucosamine sulfate were found on incident knee osteoarthritis. Nevertheless, this trial provides valuable insights for future trial design for preventive osteoarthritis studies.
Collapse
Affiliation(s)
- Jos Runhaar
- Department of General Practice, Erasmus MC University Medical Center, Rotterdam, The Netherlands.
| | - Marienke van Middelkoop
- Department of General Practice, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Max Reijman
- Department of Orthopaedics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Sten Willemsen
- Department of Biostatistics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Edwin H Oei
- Department of Radiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | | | - Gerjo van Osch
- Department of Orthopaedics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Bart Koes
- Department of General Practice, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Sita M A Bierma-Zeinstra
- Department of General Practice, Erasmus MC University Medical Center, Rotterdam, The Netherlands; Department of Orthopaedics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
133
|
Abstract
Background/Methodology Ankle arthrodesis is one popular surgical treatment for ankle arthritis, chronic instability, and degenerative deformity. However, complications such as foot pain, joint arthritis, and bone fracture may cause patients to suffer other problems. Understanding the internal biomechanics of the foot is critical for assessing the effectiveness of ankle arthrodesis and provides a baseline for the surgical plan. This study aimed to understand the biomechanical effects of ankle arthrodesis on the entire foot and ankle using finite element analyses. A three-dimensional finite element model of the foot and ankle, involving 28 bones, 103 ligaments, the plantar fascia, major muscle groups, and encapsulated soft tissue, was developed and validated. The biomechanical performances of a normal foot and a foot with ankle arthrodesis were compared at three gait instants, first-peak, mid-stance, and second-peak. Principal Findings/Conclusions Changes in plantar pressure distribution, joint contact pressure and forces, von Mises stress on bone and foot deformation were predicted. Compared with those in the normal foot, the peak plantar pressure was increased and the center of pressure moved anteriorly in the foot with ankle arthrodesis. The talonavicular joint and joints of the first to third rays in the hind- and mid-foot bore the majority of the loading and sustained substantially increased loading after ankle arthrodesis. An average contact pressure of 2.14 MPa was predicted at the talonavicular joint after surgery and the maximum variation was shown to be 80% in joints of the first ray. The contact force and pressure of the subtalar joint decreased after surgery, indicating that arthritis at this joint was not necessarily a consequence of ankle arthrodesis but rather a progression of pre-existing degenerative changes. Von Mises stress in the second and third metatarsal bones at the second-peak instant increased to 52 MPa and 34 MPa, respectively, after surgery. These variations can provide indications for outcome assessment of ankle arthrodesis surgery.
Collapse
Affiliation(s)
- Yan Wang
- Interdisciplinary Division of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University Hong Kong, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Zengyong Li
- Key Laboratory of High Efficiency and Clean Mechanical Manufacture, School of Mechanical Engineering, Shandong University, Jinan, China
| | - Duo Wai-Chi Wong
- Interdisciplinary Division of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University Hong Kong, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Ming Zhang
- Interdisciplinary Division of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University Hong Kong, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
- * E-mail:
| |
Collapse
|
134
|
Schachner ER, Lopez MJ. Diagnosis, prevention, and management of canine hip dysplasia: a review. VETERINARY MEDICINE (AUCKLAND, N.Z.) 2015; 6:181-192. [PMID: 30101105 PMCID: PMC6070021 DOI: 10.2147/vmrr.s53266] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Canine hip dysplasia (CHD) is a polygenic and multifactorial developmental disorder characterized by coxofemoral (hip) joint laxity, degeneration, and osteoarthritis (OA). Current diagnostic techniques are largely subjective measures of joint conformation performed at different stages of development. Recently, measures on three-dimensional images generated from computed tomography scans predicted the development of OA associated with CHD. Continued refinement of similar imaging methods may improve diagnostic imaging techniques to identify dogs predisposed to degenerative hip joint changes. By current consensus, joint changes consistent with CHD are influenced by genetic predisposition as well as environmental and biomechanical factors; however, despite decades of work, the relative contributions of each to the development and extent of CHD signs remain elusive. Similarly, despite considerable effort to decipher the genetic underpinnings of CHD for selective breeding programs, relevant genetic loci remain equivocal. As such, prevention of CHD within domestic canine populations is marginally successful. Conservative management is often employed to manage signs of CHD, with lifelong maintenance of body mass as one of the most promising methods. Surgical intervention is often employed to prevent joint changes or restore joint function, but there are no gold standards for either goal. To date, all CHD phenotypes are considered as a single entity in spite of recognized differences in expression and response to environmental conditions and treatment. Identification of distinct CHD phenotypes and targeting evidence-based conservative and invasive treatments for each may significantly advance prevention and management of a prevalent, debilitating condition in canine companions.
Collapse
Affiliation(s)
- Emma R Schachner
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA,
| | - Mandi J Lopez
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA,
| |
Collapse
|
135
|
Koh YG, Choi YJ, Kwon SK, Kim YS, Yeo JE. Clinical results and second-look arthroscopic findings after treatment with adipose-derived stem cells for knee osteoarthritis. Knee Surg Sports Traumatol Arthrosc 2015; 23:1308-1316. [PMID: 24326779 DOI: 10.1007/s00167-013-2807-2] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 12/01/2013] [Indexed: 02/06/2023]
Abstract
PURPOSE In the present study, the clinical outcomes and second-look arthroscopic findings of intra-articular injection of stem cells with arthroscopic lavage for treatment of elderly patients with knee osteoarthritis (OA) were evaluated. METHODS Stem cell injections combined with arthroscopic lavage were administered to 30 elderly patients (≥65 years) with knee OA. Subcutaneous adipose tissue was harvested from both buttocks by liposuction. After stromal vascular fractions were isolated, a mean of 4.04 × 10(6) stem cells (9.7 % of 4.16 × 10(7) stromal vascular fraction cells) were prepared and injected in the selected knees of patients after arthroscopic lavage. Outcome measures included the Knee Injury and Osteoarthritis Outcome Scores, visual analog scale, and Lysholm score at preoperative and 3-, 12-, and 2-year follow-up visits. Sixteen patients underwent second-look arthroscopy. RESULTS Almost all patients showed significant improvement in all clinical outcomes at the final follow-up examination. All clinical results significantly improved at 2-year follow-up compared to 12-month follow-up (P < 0.05). Among elderly patients aged >65 years, only five patients demonstrated worsening of Kellgren-Lawrence grade. On second-look arthroscopy, 87.5 % of elderly patients (14/16) improved or maintained cartilage status at least 2 years postoperatively. Moreover, none of the patients underwent total knee arthroplasty during this 2-year period. CONCLUSION Adipose-derived stem cell therapy for elderly patients with knee OA was effective in cartilage healing, reducing pain, and improving function. Therefore, adipose-derived stem cell treatment appears to be a good option for OA treatment in elderly patients. LEVEL OF EVIDENCE Therapeutic case series study, Level IV.
Collapse
Affiliation(s)
- Yong-Gon Koh
- Center for Stem Cell and Arthritis Research, Department of Orthopedic Surgery, Yonsei Sarang Hospital, 478-3, Bangbae-dong, Seocho-gu, Seoul, South Korea
| | - Yun-Jin Choi
- Center for Stem Cell and Arthritis Research, Department of Orthopedic Surgery, Yonsei Sarang Hospital, 478-3, Bangbae-dong, Seocho-gu, Seoul, South Korea.
| | - Sae-Kwang Kwon
- Center for Stem Cell and Arthritis Research, Department of Orthopedic Surgery, Yonsei Sarang Hospital, 478-3, Bangbae-dong, Seocho-gu, Seoul, South Korea
| | - Yong-Sang Kim
- Center for Stem Cell and Arthritis Research, Department of Orthopedic Surgery, Yonsei Sarang Hospital, 478-3, Bangbae-dong, Seocho-gu, Seoul, South Korea
| | - Jee-Eun Yeo
- Center for Stem Cell and Arthritis Research, Department of Orthopedic Surgery, Yonsei Sarang Hospital, 478-3, Bangbae-dong, Seocho-gu, Seoul, South Korea
| |
Collapse
|
136
|
Cipriani P, Ruscitti P, Di Benedetto P, Carubbi F, Liakouli V, Berardicurti O, Ciccia F, Triolo G, Giacomelli R. Mesenchymal stromal cells and rheumatic diseases: new tools from pathogenesis to regenerative therapies. Cytotherapy 2015; 17:832-49. [PMID: 25680301 DOI: 10.1016/j.jcyt.2014.12.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 11/24/2014] [Accepted: 12/01/2014] [Indexed: 01/08/2023]
Abstract
In recent years, mesenchymal stromal cells (MSCs) have been largely investigated and tested as a new therapeutic tool for several clinical applications, including the treatment of different rheumatic diseases. MSCs are responsible for the normal turnover and maintenance of adult mesenchymal tissues as the result of their multipotent differentiation abilities and their secretion of a variety of cytokines and growth factors. Although initially derived from bone marrow, MSCs are present in many different tissues such as many peri-articular tissues. MSCs may exert immune-modulatory properties, modulating different immune cells in both in vitro and in vivo models, and they are considered immune-privileged cells. At present, these capacities are considered the most intriguing aspect of their biology, introducing the possibility that these cells may be used as effective therapy in autoimmune diseases. Therefore, stem cell therapies may represent an innovative approach for the treatment of rheumatic diseases, especially for the forms that are not responsive to standard treatments or alternatively still lacking a definite therapy. At present, although the data from scientific literature appear to suggest that such treatments might be more effective whether administered as soon as possible, the use of MSCs in clinical practice is likely to be restricted to patients with a long history of a severe refractory disease. Further results from larger clinical trials are needed to corroborate preclinical findings and human non-controlled studies, and advancement in the knowledge of MSCs might provide information about the therapeutic role of these cells in the treatment of many rheumatic diseases.
Collapse
Affiliation(s)
- Paola Cipriani
- Rheumatology Unit, Clinical Science and Biotechnology Department, University of L'Aquila, L'Aquila, Italy.
| | - Piero Ruscitti
- Rheumatology Unit, Clinical Science and Biotechnology Department, University of L'Aquila, L'Aquila, Italy
| | - Paola Di Benedetto
- Rheumatology Unit, Clinical Science and Biotechnology Department, University of L'Aquila, L'Aquila, Italy
| | - Francesco Carubbi
- Rheumatology Unit, Clinical Science and Biotechnology Department, University of L'Aquila, L'Aquila, Italy
| | - Vasiliki Liakouli
- Rheumatology Unit, Clinical Science and Biotechnology Department, University of L'Aquila, L'Aquila, Italy
| | - Onorina Berardicurti
- Rheumatology Unit, Clinical Science and Biotechnology Department, University of L'Aquila, L'Aquila, Italy
| | - Francesco Ciccia
- Rheumatology Unit, Internal Medicine Department, University of Palermo, Palermo, Italy
| | - Giovanni Triolo
- Rheumatology Unit, Internal Medicine Department, University of Palermo, Palermo, Italy
| | - Roberto Giacomelli
- Rheumatology Unit, Clinical Science and Biotechnology Department, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
137
|
Trzeciak T, Czarny-Ratajczak M. MicroRNAs: Important Epigenetic Regulators in Osteoarthritis. Curr Genomics 2015; 15:481-4. [PMID: 25598697 PMCID: PMC4293743 DOI: 10.2174/1389202915666141024212506] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 10/14/2014] [Accepted: 10/24/2014] [Indexed: 01/17/2023] Open
Abstract
Multiple mechanisms are implicated in the development of primary osteoarthritis (OA), in which genetic and epigenetic factors appear to interact with environmental factors and age to initiate the disease and stimulate its progression. Changes in expression of microRNAs (miRs) contribute to development of osteoarthritis. Numerous miRs are involved in cartilage development, homeostasis and degradation through targeting genes expressed in this tissue. An important regulator of gene expression in human cartilage is miR-140, which directly targets a gene coding aggrecanase ADAMTS-5, that cleaves aggrecan in cartilage. This miR is considered a biological marker for cartilage and its level significantly decreases in OA cartilage. On the other hand, increased expression of miR-146a in early OA inhibits two other cartilage-degrading enzymes: MMP13 and ADAMTS4, and may provide a useful tool in developing treatments for OA. The COL2A1 gene, encoding collagen type II, which is the most abundant structural protein of the cartilage, is silenced by miR-34a and activated by miR-675. Every year, new targets of cartilage miRs are validated experimentally and this opens new possibilities for new therapies that control joint destruction and stimulate cartilage repair. At the same time development of next-generation sequencing technologies allows to identify new miRs involved in cartilage biology.
Collapse
Affiliation(s)
- Tomasz Trzeciak
- Department of Orthopedics and Traumatology, Poznan University of Medical Sciences, Poznan, Poland
| | - Malwina Czarny-Ratajczak
- Department of Medical Genetics, Poznan University of Medical Sciences, Poznan, Poland ; Department of Medicine, Center for Aging, Tulane University, School of Medicine, New Orleans, USA
| |
Collapse
|
138
|
Current perspectives in mesenchymal stem cell therapies for osteoarthritis. Stem Cells Int 2014; 2014:194318. [PMID: 25548573 PMCID: PMC4274908 DOI: 10.1155/2014/194318] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 11/19/2014] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease most commonly occurring in the ageing population. It is a slow progressive condition resulting in the destruction of hyaline cartilage followed by pain and reduced activity. Conventional treatments have little effects on the progression of the condition often leaving surgery as the last option. In the last 10 years tissue engineering utilising mesenchymal stem cells has been emerging as an alternative method for treating OA. Mesenchymal stem cells (MSCs) are multipotent progenitor cells found in various tissues, most commonly bone marrow and adipose tissue. MSCs are capable of differentiating into osteocytes, adipocytes, and chondrocytes. Autologous MSCs can be easily harvested and applied in treatment, but allogenic cells can also be employed. The early uses of MSCs focused on the implantations of cell rich matrixes during open surgeries, resulting in the formation of hyaline-like durable cartilage. More recently, the focus has completely shifted towards direct intra-articular injections where a great number of cells are suspended and injected into affected joints. In this review the history and early uses of MSCs in cartilage regeneration are reviewed and different approaches in current trends are explained and evaluated.
Collapse
|
139
|
Adams SB, Nettles DL, Jones LC, Miller SD, Guyton GP, Schon LC. Inflammatory cytokines and cellular metabolites as synovial fluid biomarkers of posttraumatic ankle arthritis. Foot Ankle Int 2014; 35:1241-9. [PMID: 25201328 DOI: 10.1177/1071100714550652] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND There is a paucity of research on posttraumatic ankle arthritis (PTAA). We aimed to identify synovial fluid PTAA biomarkers using cytokine analysis and metabolic profiling. METHODS Ankle joint synovial fluid was obtained from 20 patients with PTAA and 20 patients with no ankle pain and no radiographic evidence of ankle arthritis (control group). Synovial fluid samples were analyzed for IFN-γ, TNF-α, MIP-1β, MCP-1, IL-1β, IL-1Ra, IL-4, IL-6, IL-8, IL-10, IL-13, and IL-15 using ELISA and for more than 3000 metabolites using liquid and gas chromatography with mass spectroscopy. To compare presence of cytokines and metabolites between groups, t tests were used. Random forest analysis was performed on metabolites to determine whether control and PTAA samples could be differentiated based on metabolic profile. RESULTS IL-1Ra, IL-6, IL-8, IL-10, IL-15, and MCP-1 were significantly elevated in the PTAA group. In addition, 107 metabolites in the PTAA group were significantly altered, including derangement in amino acid, carbohydrate, lipid, and energy metabolism, extracellular matrix turnover, and collagen degradation. Random forest analysis yielded a predictive accuracy of 90% when using the metabolic profiles to distinguish between control and PTAA samples. CONCLUSION This study identified inflammatory cytokines and metabolites present in the synovial fluid of PTAA. CLINICAL RELEVANCE Several of these entities have previously been implicated in rheumatoid arthritis and osteoarthritis of the knee, but many could potentially be used as novel biomarkers of PTAA. Most importantly, the findings suggest that metabolites could be used to distinguish synovial fluid from patients with PTAA.
Collapse
Affiliation(s)
- Samuel B Adams
- Department of Orthopaedic Surgery, MedStar Union Memorial Hospital, Baltimore, MD, USA
| | - Dana L Nettles
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Lynne C Jones
- Department of Orthopaedic Surgery, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Stuart D Miller
- Department of Orthopaedic Surgery, MedStar Union Memorial Hospital, Baltimore, MD, USA
| | - Gregory P Guyton
- Department of Orthopaedic Surgery, MedStar Union Memorial Hospital, Baltimore, MD, USA
| | - Lew C Schon
- Department of Orthopaedic Surgery, MedStar Union Memorial Hospital, Baltimore, MD, USA
| |
Collapse
|
140
|
Abstract
BACKGROUND The distal interphalangeal (DIP) joints of the hand are highly susceptible to osteoarthritis and trauma. Surgical treatment options mandate accurate characterization of their osseous anatomy; however, there are few studies that describe this. We describe the curvatures of the DIP joints by measuring the bone morphology using advanced imaging and modeling methods. METHODS The fingers of 16 right hand fresh frozen human cadavers were analyzed. Fingers showing signs of DIP joint arthritis were excluded. The fingers were scanned using microtomography (microCT). Measurements of the bony morphology were made using models created from the scans. RESULTS In each finger, there is no statistically significant difference between the radii of curvature of the ulnar and radial condyles of the middle phalanx head. Conversely, the radius of curvature of the distal phalanx ulnar groove is significantly greater than that of the radial groove. The radii of curvature of the groove of the distal phalanx and the condyles of the middle phalanx displayed nonconformity with disparity increasing from the index to small fingers. Remarkably, the radius of curvature of the distal phalanx central ridge and the mean radius of the middle phalanx condyles are essentially the same. CONCLUSION The purpose of this study is to gain better insight into the DIP joints of the hand. The asymmetry between the distal phalanx grooves and the middle phalanx condyles suggests that there may be a translational component to DIP joint motion. Our understanding of morphology may lend insight into the biomechanics and disease progression within the DIP joints.
Collapse
|
141
|
Kuszel L, Trzeciak T, Richter M, Czarny-Ratajczak M. Osteoarthritis and telomere shortening. J Appl Genet 2014; 56:169-76. [PMID: 25366419 PMCID: PMC4412548 DOI: 10.1007/s13353-014-0251-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 10/04/2014] [Accepted: 10/08/2014] [Indexed: 11/16/2022]
Abstract
Osteoarthritis is the most common disease of joints caused by degradation of articular cartilage and subchondral bone. It is classified as primary form with unknown cause and as secondary form with known etiology. Genetic and epigenetic factors interact with environmental factors and contribute to the development of primary osteoarthritis. Thus far, many polymorphisms associated with osteoarthritis have been identified and recent studies also indicate the involvement of epigenetic factors (e.g., telomere shortening) in the initiation of this disorder. Accelerated shortening of telomeres was detected in osteoarthritis and other age-related diseases. Studies revealed that telomere length is severely reduced in blood leukocytes and chondrocytes of patients with osteoarthritis, and this may contribute to the initiation and development of osteoarthritis, whose major cause is still unknown.
Collapse
Affiliation(s)
- Lukasz Kuszel
- Department of Medical Genetics, Poznan University of Medical Sciences, Rokietnicka 8, 60-806, Poznan, Poland
| | | | | | | |
Collapse
|
142
|
Nieminen HJ, Salmi A, Karppinen P, Hæggström E, Hacking SA. The potential utility of high-intensity ultrasound to treat osteoarthritis. Osteoarthritis Cartilage 2014; 22:1784-99. [PMID: 25106678 DOI: 10.1016/j.joca.2014.07.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 07/22/2014] [Accepted: 07/29/2014] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) is a widespread musculoskeletal disease that reduces quality of life and for which there is no cure. The treatment of OA is challenging since cartilage impedes the local and systemic delivery of therapeutic compounds (TCs). This review identifies high-intensity ultrasound (HIU) as a non-contact technique to modify articular cartilage and subchondral bone. HIU enables new approaches to overcome challenges associated with drug delivery to cartilage and new non-invasive approaches for the treatment of joint disease. Specifically, HIU has the potential to facilitate targeted drug delivery and release deep within cartilage, to repair soft tissue damage, and to physically alter tissue structures including cartilage and bone. The localized, non-invasive ultrasonic delivery of TCs to articular cartilage and subchondral bone appears to be a promising technique in the immediate future.
Collapse
Affiliation(s)
- H J Nieminen
- Department of Physics, University of Helsinki, Finland.
| | - A Salmi
- Department of Physics, University of Helsinki, Finland.
| | - P Karppinen
- Department of Physics, University of Helsinki, Finland.
| | - E Hæggström
- Department of Physics, University of Helsinki, Finland.
| | - S A Hacking
- Department of Orthopaedics, Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
143
|
Jordan CD, Monu UD, McWalter EJ, Watkins RD, Chen W, Bangerter NK, Hargreaves BA, Gold GE. Variability of CubeQuant T1ρ, quantitative DESS T2, and cones sodium MRI in knee cartilage. Osteoarthritis Cartilage 2014; 22:1559-67. [PMID: 25278065 PMCID: PMC4185151 DOI: 10.1016/j.joca.2014.06.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 05/20/2014] [Accepted: 06/03/2014] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To measure the variability of T1ρ relaxation times using CubeQuant, T2 relaxation times using quantitative double echo in steady state (DESS), and normalized sodium signals using 3D cones sodium magnetic resonance imaging (MRI) of knee cartilage in vivo at 3 T. DESIGN Eight healthy subjects were scanned at 3 T at baseline, 1 day, 5 months, and 1 year. Ten regions of interest (ROIs) of knee cartilage were segmented in the medial and lateral compartments of each subject's knee. T1ρ and T2 relaxation times and normalized sodium signals were measured and the root-mean-square coefficient of variation (CVRMS) was calculated. Intra-subject variability was measured over short, moderate and long-term, as well as intra-observer and inter-observer variability. RESULTS The average intra-subject CVRMS measurements over short, moderate, and long-term time periods were 4.6%, 6.1%, and 6.0% for the T1ρ measurements, 6.4%, 9.3%, and 10.7% for the T2 measurements and 11.3%, 11.6%, and 12.9% for the sodium measurements, respectively. The average CVRMS measurements for intra-observer and inter-observer segmentation were 3.8% and 5.7% for the T1ρ measurements, 4.7% and 6.7% for the T2 measurements, and 8.1% and 11.4% for the sodium measurements, respectively. CONCLUSIONS These CVRMS measurements are substantially lower than previously measured changes expected in patients with advanced osteoarthritis compared to healthy volunteers, suggesting that CubeQuant T1ρ, quantitative DESS T2 and 3D cones sodium measurements are sufficiently sensitive for in vivo cartilage studies.
Collapse
Affiliation(s)
- Caroline D. Jordan
- Radiology, Stanford University, Stanford, CA, United States,Bioengineering, Stanford University, Stanford CA, United States
| | - Uchechukwuka D. Monu
- Radiology, Stanford University, Stanford, CA, United States,Electrical Engineering, Stanford University, Stanford CA, United States
| | | | | | - Weitian Chen
- GE Applied Science Laboratory, Menlo Park, CA, United States
| | - Neal K. Bangerter
- Electrical & Computer Engineering, Brigham Young University, Provo, UT, United States
| | - Brian A. Hargreaves
- Radiology, Stanford University, Stanford, CA, United States,Bioengineering, Stanford University, Stanford CA, United States,Electrical Engineering, Stanford University, Stanford CA, United States
| | - Garry E. Gold
- Radiology, Stanford University, Stanford, CA, United States,Bioengineering, Stanford University, Stanford CA, United States,Orthopaedic Surgery, Stanford University, Stanford, CA, United States
| |
Collapse
|
144
|
Quantification of differences in bone texture from plain radiographs in knees with and without osteoarthritis. Osteoarthritis Cartilage 2014; 22:1724-31. [PMID: 25278081 PMCID: PMC4587537 DOI: 10.1016/j.joca.2014.06.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 05/26/2014] [Accepted: 06/22/2014] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To quantify differences in bone texture between subjects with different stages of knee osteoarthritis (OA) and age- and gender-matched controls from plain radiographs using advanced image analysis methods. DESIGN Altogether 203 knees were imaged using constant X-ray parameters and graded according to Kellgren-Lawrence (KL) grading scale (KL0: n = 110, KL1: n = 28, KL2: n = 27, KL3: n = 31, KL4: n = 7). Bone density-related and structure-related parameters were calculated from medial and lateral tibial subchondral bone plate and trabecular bone and from femur. Density-related parameters were derived from grayscale values and structure-related parameters from Laplacian- and local binary patterns (LBP)-based images. RESULTS Reproducibilities of structure-related parameters were better than bone density-related parameters. Bone density-related parameters were significantly (P < 0.05) higher in KL2-4 groups than in control group (KL0) in medial tibial subchondral bone plate and trabecular bone. LBP-based structure parameters differed significantly between KL0 and KL2-4 groups in medial subchondral bone plate, between KL0 and KL1-4 groups in medial and lateral trabecular bone, and between KL0 and KL1-4/KL2-4 in medial and lateral femur. Laplacian-based parameters differed significantly between KL0 and KL2-4 groups in medial side regions-of-interest (ROIs). CONCLUSIONS Our results indicate that the changes in bone texture in knee OA can be quantitatively evaluated from plain radiographs using advanced image analysis. Based on the results, increased bone density can be directly estimated if the X-ray imaging conditions are constant between patients. However, structural analysis of bone was more reproducible than direct evaluation of grayscale values, and is therefore better suited for quantitative analysis when imaging conditions are variable.
Collapse
|
145
|
La Porta C, Bura SA, Negrete R, Maldonado R. Involvement of the endocannabinoid system in osteoarthritis pain. Eur J Neurosci 2014; 39:485-500. [PMID: 24494687 DOI: 10.1111/ejn.12468] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 11/25/2013] [Accepted: 12/02/2013] [Indexed: 12/29/2022]
Abstract
Osteoarthritis is a degenerative joint disease associated with articular cartilage degradation. The major clinical outcome of osteoarthritis is a complex pain state that includes both nociceptive and neuropathic mechanisms. Currently, the therapeutic approaches for osteoarthritis are limited as no drugs are available to control the disease progression and the analgesic treatment has restricted efficacy. Increasing evidence from preclinical studies supports the interest of the endocannabinoid system as an emerging therapeutic target for osteoarthritis pain. Indeed, pharmacological studies have shown the anti-nociceptive effects of cannabinoids in different rodent models of osteoarthritis, and compelling evidence suggests an active participation of the endocannabinoid system in the pathophysiology of this disease. The ubiquitous distribution of cannabinoid receptors, together with the physiological role of the endocannabinoid system in the regulation of pain, inflammation and even joint function further support the therapeutic interest of cannabinoids for osteoarthritis. However, limited clinical evidence has been provided to support this therapeutic use of cannabinoids, despite the promising preclinical data. This review summarizes the promising results that have been recently obtained in support of the therapeutic value of cannabinoids for osteoarthritis management.
Collapse
Affiliation(s)
- Carmen La Porta
- Laboratori de Neurofarmacologia, Facultat de Ciències de la Salut i de la Vida, Universitat Pompeu Fabra, C/Dr. Aiguader, 88, Barcelona, 08003, Spain
| | | | | | | |
Collapse
|
146
|
A AL, Abdel-Magied RA, El-Shereef RR, Saedii AA, AbdelGawad EA. Relationship between serum 25-hydroxy vitamin D levels, knee pain, radiological osteoarthritis, and the Western Ontario and McMaster Universities Osteoarthritis Index in patients with primary osteoarthritis. EGYPTIAN RHEUMATOLOGY AND REHABILITATION 2014. [DOI: 10.4103/1110-161x.132459] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
147
|
Lohmann-Jensen R, Holsgaard-Larsen A, Emmeluth C, Overgaard S, Jensen C. The efficacy of tourniquet assisted total knee arthroplasty on patient-reported and performance-based physical function: a randomized controlled trial protocol. BMC Musculoskelet Disord 2014; 15:110. [PMID: 24678741 PMCID: PMC3978123 DOI: 10.1186/1471-2474-15-110] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 03/20/2014] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Surgical treatment of osteoarthritis with total knee arthroplasty (TKA) usually takes place in a complete bloodless field using a tourniquet. However, doing the surgery without a tourniquet may reduce muscle damage, post-surgery pain and led to improved functional rehabilitation and mobilization. METHODS/DESIGN A prospective, blinded, parallel-group, controlled superiority trial, with balanced randomization [1:1]. Patients aged 50 or older eligible for primary TKA for osteoarthritis will be consecutively recruited from Department of Orthopedic Surgery and Traumatology, Odense University Hospital, Denmark. A total of 80 patients will be randomly allocated to TKA with or without tourniquet application providing 40 patients for each of the two treatment arms. The tourniquet assisted TKA group will have an automatic, micro-processor-based pneumatic tourniquet inflated around the thigh during surgery. The non-tourniquet assisted TKA group will have surgery performed without application of a tourniquet. The primary aim is to compare tourniquet assisted to non-tourniquet assisted TKA on patient-reported physical function (KOOS-ADL). The secondary aim is to compare post-surgery pain, function in sports and recreation, quality of life, and performance-based physical function. The explorative outcomes include; use of pain medication, single-fiber muscle damage, and changes in mechanical muscle function. The primary endpoint will be at 3-months following surgical treatment, and the time-point for analysis of the primary outcome. However, follow-up will continue up to 1 year, and provide medium-term results. The treatment effect (difference in KOOS-ADL) will be analyzed using a random effects regression model, crude and adjusted results will be reported, if needed. Analyses will be based on the intention-to-treat (ITT). Subsequent per-protocol analysis may be necessary in the event of a substantial number of patients (> 15%) being lost during follow-up. The number needed to treat (NNT) for a positive effect of treatment (>10 points on KOOS-ADL) will be reported. DISCUSSION This is the first randomized clinical trial comparing the efficacy of tourniquet assisted TKA on patient-reported physical function supported by a range of performance-based secondary outcome measures. As such it will provide high quality evidence that may help determine whether tourniquet should be used in future TKA procedures in patients with osteoarthritis of the knee. TRIAL REGISTRATION ClinicalTrials NCT01891266.
Collapse
Affiliation(s)
- Rasmus Lohmann-Jensen
- Orthopaedic Research Unit, Department of Orthopedic Surgery and Traumatology, Odense University Hospital, Odense C, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Anders Holsgaard-Larsen
- Orthopaedic Research Unit, Department of Orthopedic Surgery and Traumatology, Odense University Hospital, Odense C, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Claus Emmeluth
- Orthopaedic Research Unit, Department of Orthopedic Surgery and Traumatology, Odense University Hospital, Odense C, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Søren Overgaard
- Orthopaedic Research Unit, Department of Orthopedic Surgery and Traumatology, Odense University Hospital, Odense C, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Carsten Jensen
- Orthopaedic Research Unit, Department of Orthopedic Surgery and Traumatology, Odense University Hospital, Odense C, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense C, Denmark
| |
Collapse
|
148
|
Jang KW, Buckwalter JA, Martin JA. Inhibition of cell-matrix adhesions prevents cartilage chondrocyte death following impact injury. J Orthop Res 2014; 32:448-54. [PMID: 24249698 PMCID: PMC4034578 DOI: 10.1002/jor.22523] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 10/21/2013] [Indexed: 02/04/2023]
Abstract
Focal adhesions are transmembrane protein complexes that attach chondrocytes to the pericellular cartilage matrix and in turn, are linked to intracellular organelles via cytoskeleton. We previously found that excessive compression of articular cartilage leads to cytoskeleton-dependent chondrocyte death. Here we tested the hypothesis that this process also requires integrin activation and signaling via focal adhesion kinase (FAK) and Src family kinase (SFK). Osteochondral explants were treated with FAK and SFK inhibitors (FAKi, SFKi, respectively) for 2 h and then subjected to a death-inducing impact load. Chondrocyte viability was assessed by confocal microscopy immediately and at 24 h post-impact. With no treatment immediate post-impact viability was 59%. Treatment with 10 µM SFKi, 10 μM, or 100 µM FAKi improved viability to 80%, 77%, and 82%, respectively (p < 0.05). After 24 h viability declined to 34% in controls, 48% with 10 µM SFKi, 45% with 10 µM FAKi, and 56% with 100 µM FAKi (p < 0.01) treatment. These results confirmed that most of the acute chondrocyte mortality was FAK- and SFK-dependent, which implicates integrin-cytoskeleton interactions in the death signaling pathway. Together with previous findings, these data support the hypothesis that the excessive tissue strains accompanying impact loading induce death via a pathway initiated by strain on cell adhesion receptors.
Collapse
Affiliation(s)
- Kee W. Jang
- Department of Orthopaedics and Rehabilitation, The University of Iowa, Iowa City, IA,Department of Biomedical Engineering, The University of Iowa, Iowa City, IA
| | - Joseph A. Buckwalter
- Department of Orthopaedics and Rehabilitation, The University of Iowa, Iowa City, IA,Veterans Affairs Medical Center, Iowa City, IA
| | - James A. Martin
- Department of Orthopaedics and Rehabilitation, The University of Iowa, Iowa City, IA
| |
Collapse
|
149
|
Significant association of interleukin-4 gene intron 3 VNTR polymorphism with susceptibility to knee osteoarthritis. Gene 2014; 537:6-9. [PMID: 24406619 DOI: 10.1016/j.gene.2013.12.060] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Revised: 12/07/2013] [Accepted: 12/27/2013] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Interleukin-4 (IL-4) is a strong chondroprotective cytokine and polymorphisms within this gene may be a risk factor for osteoarthritis (OA). We aimed to investigate genotype and allele frequencies of IL-4 gene intron 3 variable number of tandem repeats (VNTR) polymorphism in patients with knee OA in a Turkish population. METHODS The study included 202 patients with knee OA and 180 healthy controls. Genomic DNA was isolated and IL-4 gene 70 bp VNTR polymorphism determined by using polymerase chain reaction (PCR) with specific primers followed by restriction fragment length polymorphism (RFLP) analysis. RESULTS Our result show that there was statistically significant difference between knee OA patients and control group with respect to IL-4 genotype distribution and allele frequencies (p=0.000, OR: 0.20, 95% CI: 0.10-0.41, OR: 0.22, 95% CI: 0.12-0.42, respectively). CONCLUSIONS Our findings suggest that there is an association of IL-4 gene intron 3 VNTR polymorphism with susceptibility of a person for development of knee OA. As a result, IL-4 gene intron 3 VNTR polymorphism could be a genetic marker in OA in a Turkish study population. This is the first association study that evaluates the associations between IL-4 gene VNTR polymorphism and knee OA.
Collapse
|
150
|
Lo KWH, Jiang T, Gagnon KA, Nelson C, Laurencin CT. Small-molecule based musculoskeletal regenerative engineering. Trends Biotechnol 2014; 32:74-81. [PMID: 24405851 DOI: 10.1016/j.tibtech.2013.12.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 12/09/2013] [Accepted: 12/10/2013] [Indexed: 01/13/2023]
Abstract
Clinicians and scientists working in the field of regenerative engineering are actively investigating a wide range of methods to promote musculoskeletal tissue regeneration. Small-molecule-mediated tissue regeneration is emerging as a promising strategy for regenerating various musculoskeletal tissues and a large number of small-molecule compounds have been recently discovered as potential bioactive molecules for musculoskeletal tissue repair and regeneration. In this review, we summarize the recent literature encompassing the past 4 years in the area of small bioactive molecules for promoting repair and regeneration of various musculoskeletal tissues including bone, muscle, cartilage, tendon, and nerve.
Collapse
Affiliation(s)
- Kevin W-H Lo
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; Department of Medicine, Division of Endocrinology, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; Department of Biomedical Engineering, University of Connecticut, School of Engineering, Storrs, CT 06268, USA.
| | - Tao Jiang
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; Department of Medicine, Division of Endocrinology, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA
| | - Keith A Gagnon
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA
| | - Clarke Nelson
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA
| | - Cato T Laurencin
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; Department of Biomedical Engineering, University of Connecticut, School of Engineering, Storrs, CT 06268, USA; Department of Orthopaedic Surgery, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; Department of Chemical, Materials and Biomolecular Engineering, University of Connecticut, School of Engineering, Storrs, CT 06268, USA.
| |
Collapse
|