49 results on '"Celeste, Scotti"'
Search Results
2. An Automatic Foot and Shank IMU Synchronization Algorithm: Proof-of-concept.
- Author
-
Shaban Shabani, Alan K. Bourke, Amir Muaremi, Jens Praestgaard, Kate O'Keeffe, Rob Argent, Martin Brom, Celeste Scotti, Brian Caulfield 0001, and Lorcan Walsh
- Published
- 2022
- Full Text
- View/download PDF
3. Test-retest reliability of wireless inertial-sensor derived measurements of knee joint kinematics.
- Author
-
Kate O'Keeffe, Rob Argent, Alan Bourke, Shaban Shabani, Jens Praestgaard, Amir Muaremi, Cathy Goulding, Martin Brom, Celeste Scotti, Lorcan Walsh, and Brian Caulfield 0001
- Published
- 2022
- Full Text
- View/download PDF
4. VEGF Over-Expression by Engineered BMSC Accelerates Functional Perfusion, Improving Tissue Density and In-Growth in Clinical-Size Osteogenic Grafts
- Author
-
Rene’ D. Largo, Maximilian G. Burger, Oliver Harschnitz, Conny F. Waschkies, Andrea Grosso, Celeste Scotti, Alexandre Kaempfen, Sinan Gueven, Gernot Jundt, Arnaud Scherberich, Dirk J. Schaefer, Andrea Banfi, and Nunzia Di Maggio
- Subjects
VEGF ,bone marrow stromal cells ,osteogenic grafts ,vascularization ,gene therapy ,Biotechnology ,TP248.13-248.65 - Abstract
The first choice for reconstruction of clinical-size bone defects consists of autologous bone flaps, which often lack the required mechanical strength and cause significant donor-site morbidity. We have previously developed biological substitutes in a rabbit model by combining bone tissue engineering and flap pre-fabrication. However, spontaneous vascularization was insufficient to ensure progenitor survival in the core of the constructs. Here, we hypothesized that increased angiogenic stimulation within constructs by exogenous VEGF can significantly accelerate early vascularization and tissue in-growth. Bone marrow stromal cells from NZW rabbits (rBMSC) were transduced with a retroviral vector to express rabbit VEGF linked to a truncated version of rabbit CD4 as a cell-surface marker. Autologous cells were seeded in clinical-size 5.5 cm3 HA scaffolds wrapped in a panniculus carnosus flap to provide an ample vascular supply, and implanted ectopically. Constructs seeded with VEGF-expressing rBMSC showed significantly increased progenitor survivival, depth of tissue ingrowth and amount of mineralized tissue. Contrast-enhanced MRI after 1 week in vivo showed significantly improved tissue perfusion in the inner layer of the grafts compared to controls. Interestingly, grafts containing VEGF-expressing rBMSC displayed a hierarchically organized functional vascular tree, composed of dense capillary networks in the inner layers connected to large-caliber feeding vessels entering the constructs at the periphery. These data constitute proof of principle that providing sustained VEGF signaling, independently of cells experiencing hypoxia, is effective to drive rapid vascularization and increase early perfusion in clinical-size osteogenic grafts, leading to improved tissue formation deeper in the constructs.
- Published
- 2020
- Full Text
- View/download PDF
5. Stem Cells for Cartilage Regeneration: A Roadmap to the Clinic
- Author
-
Celeste Scotti, Alberto Gobbi, Norimasa Nakamura, and Giuseppe M. Peretti
- Subjects
Internal medicine ,RC31-1245 - Published
- 2018
- Full Text
- View/download PDF
6. Stem Cells for Bone Regeneration: From Cell-Based Therapies to Decellularised Engineered Extracellular Matrices
- Author
-
James N. Fisher, Giuseppe M. Peretti, and Celeste Scotti
- Subjects
Internal medicine ,RC31-1245 - Abstract
Currently, autologous bone grafting represents the clinical gold standard in orthopaedic surgery. In certain cases, however, alternative techniques are required. The clinical utility of stem and stromal cells has been demonstrated for the repair and regeneration of craniomaxillofacial and long bone defects although clinical adoption of bone tissue engineering protocols has been very limited. Initial tissue engineering studies focused on the bone marrow as a source of cells for bone regeneration, and while a number of promising results continue to emerge, limitations to this technique have prompted the exploration of alternative cell sources, including adipose and muscle tissue. In this review paper we discuss the advantages and disadvantages of cell sources with a focus on adipose tissue and the bone marrow. Additionally, we highlight the relatively recent paradigm of developmental engineering, which promotes the recapitulation of naturally occurring developmental processes to allow the implant to optimally respond to endogenous cues. Finally we examine efforts to apply lessons from studies into different cell sources and developmental approaches to stimulate bone growth by use of decellularised hypertrophic cartilage templates.
- Published
- 2016
- Full Text
- View/download PDF
7. Angiopoietin-like 3-derivative LNA043 for cartilage regeneration in osteoarthritis: a randomized phase 1 trial
- Author
-
Nicole Gerwin, Celeste Scotti, Christine Halleux, Mara Fornaro, Jimmy Elliott, Yunyu Zhang, Kristen Johnson, Jian Shi, Sandra Walter, Yufei Li, Carsten Jacobi, Nelly Laplanche, Magali Belaud, Jochen Paul, Gustavo Glowacki, Thomas Peters, Keith A. Wharton, Igor Vostiar, Florine Polus, Ina Kramer, Sabine Guth, Abdelkader Seroutou, Subhajit Choudhury, Didier Laurent, Joseph Gimbel, Jörg Goldhahn, Matthias Schieker, Sophie Brachat, Ronenn Roubenoff, and Michaela Kneissel
- Subjects
General Medicine ,General Biochemistry, Genetics and Molecular Biology - Abstract
Osteoarthritis (OA) is a common, debilitating, chronic disease with no disease-modifying drug approved to date. We discovered LNA043—a derivative of angiopoietin-like 3 (ANGPTL3)—as a potent chondrogenesis inducer using a phenotypic screen with human mesenchymal stem cells. We show that LNA043 promotes chondrogenesis and cartilage matrix synthesis in vitro and regenerates hyaline articular cartilage in preclinical OA and cartilage injury models in vivo. LNA043 exerts at least part of these effects through binding to the fibronectin receptor, integrin α5β1 on mesenchymal stem cells and chondrocytes. In a first-in-human (phase 1), randomized, double-blinded, placebo-controlled, single ascending dose, single-center trial (NCT02491281; sponsored by Novartis Pharmaceuticals), 28 patients with knee OA were injected intra-articularly with LNA043 or placebo (3:1 ratio) either 2 h, 7 d or 21 d before total knee replacement. LNA043 met its primary safety endpoint and showed short serum pharmacokinetics, cartilage penetration and a lack of immunogenicity (secondary endpoints). Post-hoc transcriptomics profiling of cartilage revealed that a single LNA043 injection reverses the OA transcriptome signature over at least 21 d, inducing the expression of hyaline cartilage matrix components and anabolic signaling pathways, while suppressing mediators of OA progression. LNA043 is a novel disease-modifying OA drug candidate that is currently in a phase 2b trial (NCT04864392) in patients with knee OA.
- Published
- 2021
8. Orthotopic Bone Formation by Streamlined Engineering and Devitalization of Human Hypertrophic Cartilage
- Author
-
Sébastien, Pigeot, Paul Emile, Bourgine, Jaquiery, Claude, Celeste, Scotti, Adam, Papadimitropoulos, Atanas, Todorov, Christian, Epple, Giuseppe M, Peretti, and Ivan, Martin
- Subjects
regenerative medicine ,Mesenchymal Stem Cell Transplantation ,Article ,lcsh:Chemistry ,Osteogenesis ,Animals ,Humans ,lcsh:QH301-705.5 ,Wound Healing ,hypertrophic cartilage ,Tissue Engineering ,Tissue Scaffolds ,Skull ,apoptosis ,Cell Differentiation ,Mesenchymal Stem Cells ,bioreactors ,bone repair ,Extracellular Matrix ,Cartilage ,endochondral ossification ,lcsh:Biology (General) ,lcsh:QD1-999 ,Bone Substitutes ,Bone Remodeling ,Rabbits - Abstract
Most bones of the human body form and heal through endochondral ossification, whereby hypertrophic cartilage (HyC) is formed and subsequently remodeled into bone. We previously demonstrated that HyC can be engineered from human mesenchymal stromal cells (hMSC), and subsequently devitalized by apoptosis induction. The resulting extracellular matrix (ECM) tissue retained osteoinductive properties, leading to ectopic bone formation. In this study, we aimed at engineering and devitalizing upscaled quantities of HyC ECM within a perfusion bioreactor, followed by in vivo assessment in an orthotopic bone repair model. We hypothesized that the devitalized HyC ECM would outperform a clinical product currently used for bone reconstructive surgery. Human MSC were genetically engineered with a gene cassette enabling apoptosis induction upon addition of an adjuvant. Engineered hMSC were seeded, differentiated, and devitalized within a perfusion bioreactor. The resulting HyC ECM was subsequently implanted in a 10-mm rabbit calvarial defect model, with processed human bone (Maxgraft®, ) as control. Human MSC cultured in the perfusion bioreactor generated a homogenous HyC ECM and were efficiently induced towards apoptosis. Following six weeks of in vivo implantation, microcomputed tomography and histological analyses of the defects revealed an increased bone formation in the defects filled with HyC ECM as compared to Maxgraft®, This work demonstrates the suitability of engineered devitalized HyC ECM as a bone substitute material, with a performance superior to a state-of-the-art commercial graft. Streamlined generation of the devitalized tissue transplant within a perfusion bioreactor is relevant towards standardized and automated manufacturing of a clinical product.
- Published
- 2020
9. Monocytes Seeded on Engineered Hypertrophic Cartilage Do Not Enhance Endochondral Ossification Capacity
- Author
-
Arnaud Scherberich, Andrea Barbero, Atanas Todorov, Ivan Martin, Adam Papadimitropoulos, and Celeste Scotti
- Subjects
Adult ,Male ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Biomedical Engineering ,Mice, Nude ,Bioengineering ,Biochemistry ,Monocytes ,Biomaterials ,Glycosaminoglycan ,Extracellular matrix ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Osteogenesis ,medicine ,Animals ,Humans ,Macrophage ,Endochondral ossification ,Chemistry ,Cartilage ,Mesenchymal stem cell ,Chemotaxis ,Resorption ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Heterografts ,Female - Abstract
Engineered hypertrophic cartilage (HC) represents an attractive bone substitute material, capable to induce bone formation by endochondral ossification. Since bone formation by HC depends on factors released from the extracellular matrix, in this study, we hypothesized that HC seeding with monocytes committed to osteoclastogenesis could enhance its remodeling, improve chemotaxis of skeletal and vascular cells, and consequently enhance bone formation. This would be particularly relevant for devitalized HC, which currently exhibits only limited osteoinductivity. Living or devitalized HC engineered from human bone marrow-derived mesenchymal stromal cells (MSCs) was seeded or not with human monocytes in the presence of macrophage colony-stimulating factor and RANK-ligand, cultured for up to 15 days, or implanted ectopically in nude mice. Monocytes seeded on devitalized, but not living, HC induced its in vitro resorption, resulting in 30-fold higher release and 2.7-fold lower content of glycosaminoglycans compared with unseeded samples. In vitro, supernatants from monocyte-seeded devitalized HC attracted more monocytes compared with unseeded samples, but did not enhance chemotaxis of MSCs or human umbilical vein endothelial cells. In vivo, however, neither remodeling nor invasion by osteoclasts, endothelial cells, and mouse MSCs were significantly affected by the seeding with monocytes. Finally, in vitro priming of living or devitalized HC by monocytes did not enhance their bone-forming capacity. Further investigations should test the proposed approach on HC engineered to prevent rapid degradation and support osteoclastogenesis, or identify alternative strategies to enhance engineered HC remodeling and bone-forming capacity.
- Published
- 2017
- Full Text
- View/download PDF
10. Fat-Derived Stromal Vascular Fraction Cells Enhance the Bone-Forming Capacity of Devitalized Engineered Hypertrophic Cartilage Matrix
- Author
-
Alexander Haumer, Paul Bourgine, Matthias Kreutz, Ivan Martin, Celeste Scotti, Claude Jaquiery, Arnaud Scherberich, Andrea Barbero, and Atanas Todorov
- Subjects
Adult ,Male ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Stromal cell ,0206 medical engineering ,Mice, Nude ,Osteoclasts ,Adipose tissue ,Cell Count ,02 engineering and technology ,Choristoma ,Bone tissue ,Rats, Nude ,03 medical and health sciences ,Tissue engineering ,Osteogenesis ,Tissue Engineering and Regenerative Medicine ,medicine ,Animals ,Humans ,Cell Lineage ,Wound Healing ,Tissue Engineering ,Chemistry ,Cartilage ,Mesenchymal stem cell ,Endothelial Cells ,Hypertrophy ,Cell Biology ,General Medicine ,Anatomy ,Stromal vascular fraction ,020601 biomedical engineering ,Extracellular Matrix ,030104 developmental biology ,medicine.anatomical_structure ,Female ,Stromal Cells ,Wound healing ,Developmental Biology - Abstract
Abstract Engineered and devitalized hypertrophic cartilage (HC) has been proposed as bone substitute material, potentially combining the features of osteoinductivity, resistance to hypoxia, capacity to attract blood vessels, and customization potential for specific indications. However, in comparison with vital tissues, devitalized HC grafts have reduced efficiency of bone formation and longer remodeling times. We tested the hypothesis that freshly harvested stromal vascular fraction (SVF) cells from human adipose tissue—which include mesenchymal, endothelial, and osteoclastic progenitors—enhance devitalized HC remodeling into bone tissue. Human SVF cells isolated from abdominal lipoaspirates were characterized cytofluorimetrically. HC pellets, previously generated by human bone marrow-derived stromal cells and devitalized by freeze/thaw, were embedded in fibrin gel with or without different amounts of SVF cells and implanted either ectopically in nude mice or in 4-mm-diameter calvarial defects in nude rats. In the ectopic model, SVF cells added to devitalized HC directly contributed to endothelial, osteoblastic, and osteoclastic populations. After 12 weeks, the extent of graft vascularization and amount of bone formation increased in a cell-number-dependent fashion (up to, respectively, 2.0-fold and 2.9-fold using 12 million cells per milliliter of gel). Mineralized tissue volume correlated with the number of implanted, SVF-derived endothelial cells (CD31+ CD34+ CD146+). In the calvarial model, SVF activation of HC using 12 million cells per milliliter of gel induced efficient merging among implanted pellets and strongly enhanced (7.3-fold) de novo bone tissue formation within the defects. Our findings outline a bone augmentation strategy based on off-the-shelf devitalized allogeneic HC, intraoperatively activated with autologous SVF cells. Significance This study validates an innovative bone substitute material based on allogeneic hypertrophic cartilage that is engineered, devitalized, stored, and clinically used, together with autologous cells, intraoperatively derived from a lipoaspirate. The strategy was tested using human cells in an ectopic model and an orthotopic implantation model, in immunocompromised animals.
- Published
- 2016
- Full Text
- View/download PDF
11. Cartilage Repair in the Inflamed Joint: Considerations for Biological Augmentation Toward Tissue Regeneration
- Author
-
Norimasa Nakamura, Ivan Martin, John G. Lane, Kazunori Shimomura, Giuseppe M. Peretti, Alberto Gobbi, Georgios Karnatzikos, and Celeste Scotti
- Subjects
Cartilage, Articular ,medicine.medical_specialty ,medicine.medical_treatment ,Biomedical Engineering ,Bioengineering ,Bioinformatics ,Biochemistry ,Biomaterials ,03 medical and health sciences ,0302 clinical medicine ,Tissue engineering ,medicine ,Animals ,Humans ,Regeneration ,Autologous chondrocyte implantation ,Inflammation ,030203 arthritis & rheumatology ,Wound Healing ,030222 orthopedics ,Tissue Engineering ,business.industry ,Regeneration (biology) ,Cartilage ,Mesenchymal stem cell ,Surgery ,Clinical trial ,medicine.anatomical_structure ,Joints ,Wound healing ,business ,Adjuvant - Abstract
Cartilage repair/regeneration procedures (e.g., microfracture, autologous chondrocyte implantation [ACI]) typically result in a satisfactory outcome in selected patients. However, the vast majority of patients with chronic symptoms and, in general, a more diseased joint, do not benefit from these surgical techniques. The aims of this work were to (1) review factors negatively influencing the joint environment; (2) review current adjuvant therapies that can be used to improve results of cartilage repair/regeneration procedures in patients with more diseased joints, (3) outline future lines of research and promising experimental approaches. Chronicity of symptoms and advancing patient age appear to be the most relevant factors negatively affecting clinical outcome of cartilage repair/regeneration. Preliminary experience with hyaluronic acid, platelet-rich plasma, and mesenchymal stem cell has been positive but there is no strong evidence supporting the use of these products and this requires further assessment with high-quality, prospective clinical trials. The use of a Tissue Therapy strategy, based on more mature engineered tissues, holds promise to tackle limitations of standard ACI procedures. Current research has highlighted the need for more targeted therapies, and (1) induction of tolerance with granulocyte colony-stimulating factor (G-CSF) or by preventing IL-6 downregulation; (2) combined IL-4 and IL-10 local release; and (3) selective activation of the prostaglandin E2 (PGE2) signaling appear to be the most promising innovative strategies. For older patients and for those with chronic symptoms, adjuvant therapies are needed in combination with microfracture and ACI.
- Published
- 2016
- Full Text
- View/download PDF
12. One-step surgery with multipotent stem cells and Hyaluronan-based scaffold for the treatment of full-thickness chondral defects of the knee in patients older than 45 years
- Author
-
Alberto Gobbi, Celeste Scotti, Georgios Karnatzikos, Abhishek Mudhigere, Marc Castro, and Giuseppe M. Peretti
- Subjects
Scaffold ,Bone Marrow Aspirate Concentrate (BMAC) ,030222 orthopedics ,03 medical and health sciences ,Cartilage ,0302 clinical medicine ,Cartilage lesion ,Knee ,Orthopedics and Sports Medicine ,Surgery ,030229 sport sciences ,Mesenchymal Stem cells (MSCs) ,Hyaluronan - Abstract
Purpose The aim of this study is to prospectively evaluate the medium-term effectiveness and regenerative capability of autologous adult mesenchymal stem cells, harvested as bone marrow aspirate concentrate (BMAC), along with a hyaluronan-based scaffold (Hyalofast) in the treatment of ICRS grade 4 chondral lesions of the knee joint, in patients older than 45 years. Methods A study group of 20 patients with an age >45 years (mean 50.0 ± 4.1 years) was compared to a control group of 20 patients with an age 45 years population with functional outcomes that are comparable to younger patients at final follow-up. Level of evidence Prospective cohort study, Level II.
- Published
- 2016
- Full Text
- View/download PDF
13. Thoracic Outlet Syndrome in the Overhead Athlete
- Author
-
Giuseppe M. Peretti, Simonetta Gerevini, Celeste Scotti, M Agnoletto, Gianfranco Fraschini, R. Ballis, and Pietro Ciampi
- Subjects
Adult ,Male ,0301 basic medicine ,Thoracic outlet ,Weakness ,medicine.medical_specialty ,Pain ,Physical Therapy, Sports Therapy and Rehabilitation ,Pectoralis Muscles ,Muscle hypertrophy ,Young Adult ,03 medical and health sciences ,medicine ,Humans ,Orthopedics and Sports Medicine ,Supernumerary ,Pectoralis Muscle ,Thoracic outlet syndrome ,business.industry ,medicine.disease ,Surgery ,body regions ,Thoracic Outlet Syndrome ,medicine.anatomical_structure ,Athletic Injuries ,Subclavius muscle ,Upper limb ,030101 anatomy & morphology ,medicine.symptom ,business ,human activities ,Magnetic Resonance Angiography - Abstract
Subclavius posticus muscle is a supernumerary anatomical variation of the subclavius muscle. The aim of this study was to show the possible contribution of the posticus muscle in the development of unilateral thoracic outlet syndrome (TOS) in overhead athletes, presenting hypertrophy of the dominant arm due to their sport activity. Reported here are 2 young overhead athletes complaining pain, paresthesia, weakness in the dominant upper limb, although presenting none of the main shoulder and neurological disorders. After developing subclavian vein thrombosis, TOS was suspected and finally diagnosed by dynamic magnetic resonance angiography, which also showed bilateral subclavius posticus muscle in both patients. Despite bilateral subclavius posticus, TOS was only evident in the dominant limb in which the association of hypertrophy of lateral cervical muscles, resulting from the overhead sport activity, to the subclavius posticus likely led to a significant reduction of the upper thoracic outlet space.
- Published
- 2017
- Full Text
- View/download PDF
14. The comparison of the performance of 3 T and 7 T T
- Author
-
Vladimir, Juras, Markus, Schreiner, Didier, Laurent, Štefan, Zbýň, Vladimir, Mlynarik, Pavol, Szomolanyi, Benedikt, Hager, Celeste, Scotti, Jörg, Goldhahn, Rahel, Heule, Oliver, Bieri, and Siegfried, Trattnig
- Subjects
Adult ,Cartilage, Articular ,Male ,Observer Variation ,Knee Joint ,Tibia ,Reproducibility of Results ,Patella ,Middle Aged ,Magnetic Resonance Imaging ,Article ,Image Processing, Computer-Assisted ,Humans ,Female ,Femur - Abstract
OBJECTIVE: To investigate T(2) mapping as a possible marker for low-grade human articular cartilage lesions during a one-year follow-up, possible changes during the follow-up and compare the reliability and sensitivity of these measurements on high-field (3 T) and ultra-high-field (7 T) MRI scanners. DESIGN: Twenty-one patients with femoral, tibial and patellar cartilage defect in the knee joint participated in the study. The MRI protocol consisted of morphological, as well as three-dimensional triple-echo steady-state (3D-TESS) T(2) mapping sequences with similar parameters at 3T and 7T. Patients were scanned at five time-points up to 12 months. T(2) values were evaluated in the lesion and healthy-appearing regions for superficial and deep cartilage zone. The repeated ANOVA was used to determine differences in T(2) values at various time points. RESULTS: A significant decrease in T(2) values was observed between baseline and six months in the superficial layer of the lesion in patients at 3 T (decrease from 41.89 ± 9.3 ms to 31.21 ± 7.2 ms, which is a difference of −5.67 ± 2.2 ms (p = 0.031)), and at 12 months in the superficial layer of the lesion in patients at 3 T (decrease from 41.89 ± 9.3 ms to 35.28 ± 4.9 ms, which is a difference of −6.60 ± 4.4 ms (p = 0.044). No significant differences were recorded at 7 T. CONCLUSION: The change in T(2) values acquired with 3 T 3D-TESS appears to be reflecting subtle changes of cartilage composition in the course of low-grade lesion development. 7 T T(2) mapping does not reflect these changes probably due to completely decayed short T(2) component.
- Published
- 2018
15. Generation and characterization of osteochondral grafts with human nasal chondrocytes
- Author
-
Michele Pansini, Dieter Wirz, Ivan Martin, Oliver Bieri, Daniel Baumhoer, Andrea Barbero, Marina Barandun, Lukas Daniel Iselin, Ueli Studler, Dirk J. Schaefer, Francesco Santini, Martin Haug, Celeste Scotti, and Marcel Jakob
- Subjects
Glycosaminoglycan ,medicine.anatomical_structure ,Tissue engineering ,Chemistry ,Cartilage ,Engineered cartilage ,medicine ,Orthopedics and Sports Medicine ,Bone matrix ,Anatomy ,Biomedical engineering - Abstract
We investigated whether nasal chondrocytes (NC) can be used to generate composite constructs with properties necessary for the repair of osteochondral (OC) lesions, namely maturation, integration and capacity to recover from inflammatory burst. OC grafts were fabricated by combining engineered cartilage tissues (generated by culturing NC or articular chondrocytes - AC - onto Chondro-Gide® matrices) with devitalized spongiosa cylinders (Tutobone®). OC tissues were then exposed to IL-1β for three days and cultured for additional 2 weeks in the absence of IL-1β. Cartilage maturation extent was assessed (immune) histologically, biochemically and by delayed gadolinium-enhanced magnetic resonance imaging of cartilage (dGEMRIC) while cartilage/bone integration was assessed using a peel-off mechanical test. The use of NC as compared to AC allowed for more efficient cartilage matrix accumulation and superior integration of the cartilage/bone layers. dGEMRIC and biochemical analyzes of the OC constructs showed a reduced glycosaminoglycan (GAG) contents upon IL-1β administration. Cartilaginous matrix contents and integration forces returned to baseline up on withdrawal of IL-1β. By having a cartilage layer well developed and strongly integrated to the subchondral layer, OC tissues generated with NC may successfully engraft in an inflammatory post-surgery joint environment.
- Published
- 2015
- Full Text
- View/download PDF
16. Engineered decellularized matrices to instruct bone regeneration processes
- Author
-
Adam Papadimitropoulos, Paul Bourgine, Ivan Martin, Arnaud Scherberich, and Celeste Scotti
- Subjects
Bone Regeneration ,Histology ,Physiology ,Computer science ,Endocrinology, Diabetes and Metabolism ,02 engineering and technology ,Bone healing ,Bone tissue ,Extracellular matrix ,03 medical and health sciences ,Tissue engineering ,medicine ,Animals ,Humans ,Progenitor cell ,Bone regeneration ,030304 developmental biology ,Wound Healing ,0303 health sciences ,Decellularization ,Tissue Engineering ,Anatomy ,Reference Standards ,021001 nanoscience & nanotechnology ,Extracellular Matrix ,medicine.anatomical_structure ,Stem cell ,0210 nano-technology ,Neuroscience - Abstract
Despite the significant progress in the field of bone tissue engineering, cell-based products have not yet reached the stage of clinical adoption. This is due to the uncertain advantages from the standard-of-care, combined with challenging cost-and regulatory-related issues. Novel therapeutic approaches could be based on exploitation of the intrinsic regenerative capacity of bone tissue, provided the development of a deeper understanding of its healing mechanisms. While it is well-established that endogenous progenitors can be activated toward bone formation by overdoses of single morphogens, the challenge to stimulate the healing processes by coordinated and controlled stimulation of specific cell populations remains open. Here, we review the recent approaches to generate osteoinductive materials based on the use of decellularized extracellular matrices (ECM) as reservoirs of multiple factors presented at physiological doses and through the appropriate ligands. We then propose the generation of customized engineered and decellularized ECM (i) as a tool to better understand the processes of bone regeneration and (ii) as safe and effective "off-the-shelf" bone grafts for clinical use. This article is part of a Special Issue entitled Stem Cells and Bone.
- Published
- 2015
- Full Text
- View/download PDF
17. Meniscus repair and regeneration: review on current methods and research potential
- Author
-
Giuseppe M. Peretti, Pierluigi Antinolfi, Michael T. Hirschmann, Celeste Scotti, and Ivan Martin
- Subjects
Scaffold ,lcsh:Diseases of the musculoskeletal system ,lcsh:Surgery ,Meniscal tears ,scaffold ,Meniscus (anatomy) ,Mesenchymal Stem Cell Transplantation ,Regenerative Medicine ,Menisci, Tibial ,Regenerative medicine ,bioreactor ,Tissue engineering ,medicine ,Animals ,Humans ,Regeneration ,Meniscus ,Meniscus repair ,meniscal tear ,Regeneration (biology) ,lcsh:RD1-811 ,musculoskeletal system ,medicine.anatomical_structure ,tissue engineering ,Fibrocartilage ,Joint Diseases ,lcsh:RC925-935 ,Biomedical engineering - Abstract
Meniscus regeneration is an unsolved clinical challenge. Despite the wide acceptance of the degenerative consequences of meniscectomy, no surgical procedure has succeeded to date in regenerating a functional and long-lasting meniscal fibrocartilage. Research proposed a number of experimental approaches encompassing all the typical strategies of regenerative medicine: cell-free scaffolds, gene therapy, intra-articular delivery of progenitor cells, biological glues for enhanced bonding of reparable tears, partial and total tissue engineered meniscus replacement. None of these approaches has been completely successful and can be considered suitable for all patients, as meniscal tears require specific and patient-related treatments depending on the size and type of lesion. Recent advances in cell biology, biomaterial science and bioengineering (e.g., bioreactors) have now the potential to drive meniscus regeneration into a series of clinically relevant strategies. In this tutorial paper, the clinical need for meniscus regeneration strategies will be explained, and past and current experimental studies on meniscus regeneration will be reported.
- Published
- 2013
- Full Text
- View/download PDF
18. Scaffold-Based Delivery of a Clinically Relevant Anti-Angiogenic Drug Promotes the Formation of In Vivo Stable Cartilage
- Author
-
Franca Abbruzzese, Marcella Trombetta, Matteo Centola, Andrea Barbero, Alberto Rainer, Anna Marsano, Vincenzo Denaro, Gianluca Vadalà, Ivan Martin, and Celeste Scotti
- Subjects
Adult ,Male ,Vascular Endothelial Growth Factor A ,Scaffold ,Angiogenesis ,Biomedical Engineering ,Angiogenesis Inhibitors ,Enzyme-Linked Immunosorbent Assay ,Bioengineering ,Antibodies, Monoclonal, Humanized ,Real-Time Polymerase Chain Reaction ,Biochemistry ,Biomaterials ,Cell therapy ,Mice ,chemistry.chemical_compound ,Tissue engineering ,Cell Movement ,In vivo ,Hyaluronic acid ,Human Umbilical Vein Endothelial Cells ,medicine ,Animals ,Humans ,Hyaluronic Acid ,Autologous chondrocyte implantation ,Aged ,Aged, 80 and over ,Fibrin ,Tissue Engineering ,Tissue Scaffolds ,Reverse Transcriptase Polymerase Chain Reaction ,business.industry ,Cartilage ,Original Articles ,Middle Aged ,Bevacizumab ,medicine.anatomical_structure ,chemistry ,Immunology ,Microscopy, Electron, Scanning ,Cancer research ,Female ,business - Abstract
Standard cartilage tissue engineering approaches, for example, matrix-induced autologous chondrocyte implantation (MACI), consist of the implantation of cell-based constructs whose survival and further development first depend on the degree of graft maturity at the time of surgery (e.g., matrix production) and, subsequently, on initial host reaction. Indeed, blood vessel ingrowth and macrophage migration within the implant may endanger graft stability of immature constructs; so, control of angiogenesis was proposed as an adjuvant of cellular therapy for the treatment of cartilage defects. In this study, we hypothesized that engineered constructs with no in vitro precultivation, but functionalized to block angiogenesis right on implantation, might result in better survival, as well as superior long-term cartilaginous quality. Here, we propose a clinically compatible fibrin/hyaluronan scaffold seeded with nasal chondrocytes (NC) and functionalized with an FDA-approved anti-angiogenic drug (bevacizumab; Avastin(®)), which sequestrates vascular endothelial growth factor from the surrounding environment. Our results show that the sustained bevacizumab release from NC-loaded scaffolds after subcutaneous implantation in nude mice efficiently blocked host vessels ingrowth (five times lower CD31(+) cells infiltration vs. control group, at 3 weeks after implant), and enhanced constructs survival rate (75% vs. 18% for the control, at 6 weeks after implant). In vitro assays, developed to elucidate the role of specific construct components in the in vivo remodeling, allowed to determine that fibrin degradation products enhanced the in vitro endothelial cell proliferation, as well as the macrophage migration; whereas the presence of bevacizumab was capable of counteracting these effects. The proposed pharmacological control of angiogenesis by a therapeutic drug released from a scaffold might enhance cartilage regeneration by MACI approaches, possibly allowing it to bypass the complex and costly phase of graft preculture to gain increased functionality.
- Published
- 2013
- Full Text
- View/download PDF
19. Animal models for meniscus repair and regeneration
- Author
-
Giuseppe M. Peretti, Ivan Martin, Celeste Scotti, D. Deponti, and Alessia Di Giancamillo
- Subjects
Knee function ,Engineering ,business.industry ,Regeneration (biology) ,Biomedical Engineering ,Medicine (miscellaneous) ,Meniscus (anatomy) ,musculoskeletal system ,Regenerative medicine ,Biomaterials ,Knee meniscus ,medicine.anatomical_structure ,Basic research ,medicine ,Gross anatomy ,business ,Meniscus repair ,Biomedical engineering - Abstract
The meniscus plays an important role in knee function and mechanics. Meniscal lesions, however, are common phenomena and this tissue is not able to achieve spontaneous successful repair, particularly in the inner avascular zone. Several animal models have been studied and proposed for testing different reparative approaches, as well as for studying regenerative methods aiming to restore the original shape and function of this structure. This review summarizes the gross anatomy, function, ultrastructure and biochemical composition of the knee meniscus in several animal models in comparison with the human meniscus. The relevance of the models is discussed from the point of view of basic research as well as of clinical translation for meniscal repair, substitution and regeneration. Finally, the advantages and disadvantages of each model for various research directions are critically discussed.
- Published
- 2013
- Full Text
- View/download PDF
20. Engineering Small-Scale and Scaffold-Based Bone Organs via Endochondral Ossification Using Adult Progenitor Cells
- Author
-
Celeste, Scotti, Beatrice, Tonnarelli, Adam, Papadimitropoulos, Elia, Piccinini, Atanas, Todorov, Matteo, Centola, Andrea, Barbero, and Ivan, Martin
- Subjects
Adult ,Bone Transplantation ,Tissue Engineering ,Tissue Scaffolds ,Cell Culture Techniques ,Mesenchymal Stem Cells ,Adult Stem Cells ,Mice ,Cartilage ,Osteogenesis ,Animals ,Humans ,Diastasis, Bone ,Cells, Cultured - Abstract
Bone development, growth, and repair predominantly occur through the process of endochondral ossification, characterized by remodelling of cartilaginous templates. The same route efficiently supports engineering of bone marrow as a niche for hematopoietic stem cells (HSC). Here we describe a combined in vitro/in vivo system based on bone marrow-derived Mesenchymal Stem/Stromal Cells (MSC) that duplicates the hallmark cellular and molecular events of endochondral ossification during development. The model requires MSC culture with instructive molecules to generate hypertrophic cartilage tissues. The resulting constructs complete the endochondral route upon in vivo implantation, in the timeframe of up to 12 weeks. The described protocol is clearly distinct from the direct ossification approach typically used to drive MSC towards osteogenesis. Recapitulation of endochondral ossification allows modelling of stromal-HSC interactions in physiology and pathology and allows engineering processes underlying bone regeneration.
- Published
- 2016
21. The Benefit of Synthetic Versus Biological Patch Augmentation in the Repair of Posterosuperior Massive Rotator Cuff Tears: A 3-Year Follow-up Study
- Author
-
Giuseppe M. Peretti, Matteo Vitali, Pietro Ciampi, Celeste Scotti, Gianfranco Fraschini, Alessandro Nonis, Clelia Di Serio, Pietro, Ciampi, Celeste, Scotti, Alessandro, Noni, Matteo, Vitali, DI SERIO, Mariaclelia, Giuseppe M., Peretti, and Gianfranco, Fraschini
- Subjects
Male ,medicine.medical_specialty ,Visual Analog Scale ,Visual analogue scale ,Physical Therapy, Sports Therapy and Rehabilitation ,Polypropylenes ,Rotator Cuff Injuries ,Rotator Cuff ,Suture (anatomy) ,Recurrence ,Materials Testing ,medicine ,Humans ,Orthopedics and Sports Medicine ,Rotator cuff ,Muscle Strength ,Aged ,Retrospective Studies ,Bioprosthesis ,Rupture ,business.industry ,Follow up studies ,Age Factors ,Middle Aged ,Surgical Mesh ,Tendon ,Surgery ,medicine.anatomical_structure ,Case-Control Studies ,Linear Models ,Open repair ,Tears ,Female ,Collagen ,business ,Pericardium ,Cohort study ,Follow-Up Studies - Abstract
Background:Rotator cuff repair typically results in a satisfactory, although variable, clinical outcome. However, anatomic failure of the repaired tendon often occurs.Hypothesis:Patch augmentation can improve the results of open rotator cuff repair by supporting the healing process, protecting the suture, and reducing friction in the subacromial space.Study Design:Cohort study; Level of evidence, 3.Methods:A total of 152 patients with a posterosuperior massive rotator cuff tear were treated by open repair only (control group; n = 51; mean age, 67.06 ± 4.42 years), open repair together with collagen patch augmentation (collagen group; n = 49; mean age, 66.53 ± 5.17 years), or open repair together with polypropylene patch augmentation (polypropylene group; n = 52; mean age, 66.17 ± 5.44 years) and were retrospectively studied. Patients were evaluated preoperatively and after 36 months with a visual analog scale (VAS) and the University of California, Los Angeles (UCLA) shoulder rating scale and by measuring elevation of the scapular plane and strength with a dynamometer. The VAS and UCLA scores were also obtained 2 months postoperatively. Tendon integrity was assessed after 1 year by ultrasound. Patients were homogeneous as per the preoperative assessment.Results:After 2 months, results (mean ± standard deviation) for the control, collagen, and polypropylene groups, respectively, were as follows: VAS scores were 6.96 ± 1.11, 6.46 ± 1.02, and 4.92 ± 0.90, while UCLA scores were 11.29 ± 1.46, 11.40 ± 1.51, and 19.15 ± 1.99. After 36 months, the mean scores for the respective groups were 3.66 ± 1.05, 4.06 ± 1.02, and 3.28 ± 1.10 for the VAS and 14.88 ± 1.98, 14.69 ± 1.99, and 24.61 ± 3.22 for the UCLA scale. In addition, after 36 months, elevation on the scapular plane was 140.68° ± 9.84°, 140.61° ± 12.48°, and 174.71° ± 8.18°, and abduction strength was 8.73 ± 0.54 kg, 9.03 ± 0.60 kg, and 13.79 ± 0.64 kg for the control, collagen, and polypropylene groups, respectively. The retear rate after 12 months was 41% (21/51) for the control group, 51% (25/49) for the collagen group, and 17% (9/52) for the polypropylene group. In particular, the reduced 12-month retear rate and the increased UCLA scores, abduction strength, and elevation at 3-year follow-up were statistically significant for patients treated with a polypropylene patch compared with those treated with repair only or with a collagen patch.Conclusion:Polypropylene patch augmentation of rotator cuff repair was demonstrated to significantly improve the 36-month outcome in terms of function, strength, and retear rate.
- Published
- 2014
22. Scaffolding as Treatment for Osteochondral Defects in the Ankle
- Author
-
Giuseppe M. Peretti, Celeste Scotti, and Alberto Gobbi
- Subjects
medicine.medical_specialty ,Scaffold ,business.industry ,Mesenchymal stem cell ,Osteoarthritis ,medicine.disease ,Osteochondritis dissecans ,Regenerative medicine ,Surgery ,medicine.anatomical_structure ,Tissue engineering ,Orthopedic surgery ,medicine ,Ankle ,business - Abstract
Osteochondral lesions of the talus can occur after ankle trauma (e.g., inversion sprains) or be idiopathic, in case of osteochondritis dissecans. In both scenarios, it is challenging for the orthopedic surgeon to establish the optimal therapy. Treatment options include both conservative and surgical procedures depending on severity, size, and location of the lesion. The aim of surgery is to restore the articular surface and, therefore, possibly avoid or delay the onset of osteoarthritis. Thanks to the recent advancements in tissue engineering and regenerative medicine, scaffold-based techniques, either cell-free or cell based, have been proposed for cartilage repair in the ankle. The optimal scaffold should simultaneously promote high rates of cell division and cartilaginous extracellular matrix synthesis by phenotypically stable chondrocytes. In this chapter, general characteristics of scaffolds and the main scaffolds and procedures used for cartilage repair in the ankle will be discussed.
- Published
- 2016
- Full Text
- View/download PDF
23. Stem Cells for Bone Regeneration: From Cell-Based Therapies to Decellularised Engineered Extracellular Matrices
- Author
-
Celeste Scotti, Giuseppe M. Peretti, and James N. Fisher
- Subjects
0301 basic medicine ,Bone growth ,lcsh:Internal medicine ,Stromal cell ,business.industry ,Regeneration (biology) ,Long bone ,Cell Biology ,Review Article ,Bioinformatics ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Tissue engineering ,Medicine ,Bone marrow ,Stem cell ,business ,Bone regeneration ,lcsh:RC31-1245 ,Molecular Biology - Abstract
Currently, autologous bone grafting represents the clinical gold standard in orthopaedic surgery. In certain cases, however, alternative techniques are required. The clinical utility of stem and stromal cells has been demonstrated for the repair and regeneration of craniomaxillofacial and long bone defects although clinical adoption of bone tissue engineering protocols has been very limited. Initial tissue engineering studies focused on the bone marrow as a source of cells for bone regeneration, and while a number of promising results continue to emerge, limitations to this technique have prompted the exploration of alternative cell sources, including adipose and muscle tissue. In this review paper we discuss the advantages and disadvantages of cell sources with a focus on adipose tissue and the bone marrow. Additionally, we highlight the relatively recent paradigm of developmental engineering, which promotes the recapitulation of naturally occurring developmental processes to allow the implant to optimally respond to endogenous cues. Finally we examine efforts to apply lessons from studies into different cell sources and developmental approaches to stimulate bone growth by use of decellularised hypertrophic cartilage templates.
- Published
- 2016
24. Engineering Small-Scale and Scaffold-Based Bone Organs via Endochondral Ossification Using Adult Progenitor Cells
- Author
-
Andrea Barbero, Atanas Todorov, Beatrice Tonnarelli, Celeste Scotti, Elia Piccinini, Ivan Martin, Matteo Centola, and Adam Papadimitropoulos
- Subjects
0301 basic medicine ,Stromal cell ,Mesenchymal stem cell ,Anatomy ,Biology ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Intramembranous ossification ,medicine ,Direct ossification ,Bone marrow ,Stem cell ,Bone regeneration ,Endochondral ossification - Abstract
Bone development, growth, and repair predominantly occur through the process of endochondral ossification, characterized by remodelling of cartilaginous templates. The same route efficiently supports engineering of bone marrow as a niche for hematopoietic stem cells (HSC). Here we describe a combined in vitro/in vivo system based on bone marrow-derived Mesenchymal Stem/Stromal Cells (MSC) that duplicates the hallmark cellular and molecular events of endochondral ossification during development. The model requires MSC culture with instructive molecules to generate hypertrophic cartilage tissues. The resulting constructs complete the endochondral route upon in vivo implantation, in the timeframe of up to 12 weeks. The described protocol is clearly distinct from the direct ossification approach typically used to drive MSC towards osteogenesis. Recapitulation of endochondral ossification allows modelling of stromal-HSC interactions in physiology and pathology and allows engineering processes underlying bone regeneration.
- Published
- 2016
- Full Text
- View/download PDF
25. Interleukin-1β modulates endochondral ossification by human adult bone marrow stromal cells
- Author
-
Marcus Mumme, Adam Papadimitropoulos, David Wendt, Andrea Barbero, Chiara Bocelli-Tyndall, Marcel Jakob, Atanas Todorov, Celeste Scotti, Waldemar Hoffmann, and Ivan Martin
- Subjects
Adult ,Male ,lcsh:Diseases of the musculoskeletal system ,Stromal cell ,Interleukin-1beta ,lcsh:Surgery ,Bone Morphogenetic Protein 2 ,Bone healing ,osteogenesis ,Mice ,Matrix Metalloproteinase 13 ,chondrogenesis ,Bone cell ,medicine ,Animals ,Humans ,RNA, Messenger ,Endochondral ossification ,Cells, Cultured ,Cell Proliferation ,Glycosaminoglycans ,Osteoblasts ,Chemistry ,Cartilage ,Cell Differentiation ,lcsh:RD1-811 ,Anatomy ,Fibroblasts ,Middle Aged ,Chondrogenesis ,Cell biology ,endochondral ossification ,medicine.anatomical_structure ,tissue engineering ,Intramembranous ossification ,Mesenchymal stem cells ,Calcium ,Bone marrow ,lcsh:RC925-935 - Abstract
Inflammatory cytokines present in the milieu of the fracture site are important modulators of bone healing. Here we investigated the effects of interleukin-1β (IL-1β) on the main events of endochondral bone formation by human bone marrow mesenchymal stromal cells (BM-MSC), namely cell proliferation, differentiation and maturation/remodelling of the resulting hypertrophic cartilage. Low doses of IL-1β (50 pg/mL) enhanced colony-forming units-fibroblastic (CFU-f) and -osteoblastic (CFU-o) number (up to 1.5-fold) and size (1.2-fold) in the absence of further supplements and glycosaminoglycan accumulation (1.4-fold) upon BM-MSC chondrogenic induction. In osteogenically cultured BM-MSC, IL-1β enhanced calcium deposition (62.2-fold) and BMP-2 mRNA expression by differential activation of NF-κB and ERK signalling. IL-1β-treatment of BM-MSC generated cartilage resulted in higher production of MMP-13 (14.0-fold) in vitro, mirrored by an increased accumulation of the cryptic cleaved fragment of aggrecan, and more efficient cartilage remodelling/resorption after 5 weeks in vivo (i.e., more TRAP positive cells and bone marrow, less cartilaginous areas), resulting in the formation of mature bone and bone marrow after 12 weeks. In conclusion, IL-1β finely modulates early and late events of the endochondral bone formation by BM-MSC. Controlling the inflammatory environment could enhance the success of therapeutic approaches for the treatment of fractures by resident MSC and as well as improve the engineering of implantable tissues.
- Published
- 2012
- Full Text
- View/download PDF
26. Autologous Tissue-engineered Osteochondral Graft for Talus Osteochondral Lesions
- Author
-
Andrea Barbero, Dirk J. Schaefer, Ivan Martin, Christian Candrian, Celeste Scotti, Davide Croci, André Leumann, Marcel Jakob, and Victor Valderrabano
- Subjects
Scaffold ,Tissue engineering ,business.industry ,Medicine ,Biomaterial ,Orthopedics and Sports Medicine ,Surgery ,business ,Autologous tissue ,Biomedical engineering - Published
- 2011
- Full Text
- View/download PDF
27. Posterior spinal instrumentation: biomechanical study on the role of rods on hardware response to axial load
- Author
-
Giuseppe Sala, Davide Mandelli, Giuseppe Gioia, and Celeste Scotti
- Subjects
medicine.medical_treatment ,Posterior spinal instrumentation ,Lumbar vertebrae ,medicine.disease_cause ,Weight-bearing ,Weight-Bearing ,Materials Testing ,medicine ,Humans ,Biomechanics ,Orthopedics and Sports Medicine ,Reduction (orthopedic surgery) ,Lumbar Vertebrae ,Dynamic stabilization ,In vitro testing ,business.industry ,Work (physics) ,Process (computing) ,Stiffness ,Prostheses and Implants ,Biomechanical Phenomena ,Spinal Fusion ,medicine.anatomical_structure ,Spinal fusion ,Original Article ,Surgery ,medicine.symptom ,business ,Computer hardware - Abstract
Posterior spinal instrumentation is frequently used for the treatment of spine disorders. Importantly, different requirements have to be considered for the optimal use of these systems in various clinical scenarios. In this work, we focused on the role of rods diameter on hardware's stiffness. For this purpose, we established an in vitro model and compared the response to axial load of a posterior stabilization system, characterized by rods of different diameter (4, 5, 6 mm), with that of Dynesys®. Intuitively, the higher the stiffness of the hardware, the lower the load is transferred to the disc. However, the 4 hardware tested showed a different trend in the response to the load regimens: when increasing the load, more flexible systems display a progressive reduction in the percentage of load which is transferred to the disc while more rigid system display the opposite trend. Considering that the load which is transferred, and not by-passed by the hardware, influences the healing of a fracture; the integration of a bone graft or a cage; the fusion process, these data have a relevant impact on clinical practice and highlight features that have to be considered in the choice for the optimal posterior spinal instrumentation.
- Published
- 2011
- Full Text
- View/download PDF
28. One-Step Cartilage Repair with Bone Marrow Aspirate Concentrated Cells and Collagen Matrix in Full-Thickness Knee Cartilage Lesions
- Author
-
Laura Mazzucco, Vivek Mahajan, Brunella Grigolo, Celeste Scotti, Alberto Gobbi, and Georgios Karnatzikos
- Subjects
Collagen i ,medicine.medical_specialty ,Pathology ,business.industry ,Cartilage ,Biomedical Engineering ,Physical Therapy, Sports Therapy and Rehabilitation ,Matrix (biology) ,Surgery ,Knee cartilage ,Bone marrow aspirate ,medicine.anatomical_structure ,medicine ,Immunology and Allergy ,Full thickness ,business ,Cartilage repair - Abstract
Objective: The purpose of our study was to determine the effectiveness of cartilage repair utilizing 1-step surgery with bone marrow aspirate concentrate (BMAC) and a collagen I/III matrix (Chondro-Gide, Geistlich, Wolhusen, Switzerland). Materials and Methods: We prospectively followed up for 2 years 15 patients (mean age, 48 years) who were operated for grade IV cartilage lesions of the knee. Six of the patients had multiple chondral lesions; the average size of the lesions was 9.2 cm2. All patients underwent a mini-arthrotomy and concomitant transplantation with BMAC covered with the collagen matrix. Coexisting pathologies were treated before or during the same surgery. X-rays and MRI were collected preoperatively and at 1 and 2 years’ follow-up. Visual analog scale (VAS), International Knee Documentation Committee (IKDC), Knee injury and Osteoarthritis Outcome Score (KOOS), Lysholm, Marx, SF-36 (physical/mental), and Tegner scores were collected preoperatively and at 6, 12, and 24 months’ follow-up. Four patients gave their consent for second-look arthroscopy and 3 of them for a concomitant biopsy. Results: Patients showed significant improvement in all scores at final follow-up ( P < 0.005). Patients presenting single lesions and patients with small lesions showed higher improvement. MRI showed coverage of the lesion with hyaline-like tissue in all patients in accordance with clinical results. Hyaline-like histological findings were also reported for all the specimens analyzed. No adverse reactions or postoperative complications were noted. Conclusion: This study showed that 1-step surgery with BMAC and collagen I/III matrix could be a viable technique in the treatment of grade IV knee chondral lesions.
- Published
- 2011
- Full Text
- View/download PDF
29. Blood exposure has a negative effect on engineered cartilage
- Author
-
Antonio Gigante, Giuseppe M. Peretti, S Manzotti, C. Sosio, S. Biressi, Laura Mangiavini, Federica Boschetti, Gianfranco Fraschini, Celeste Scotti, and M. S. Buragas
- Subjects
Cartilage, Articular ,Scaffold ,Swine ,Chondrocyte ,Chondrocytes ,Tissue engineering ,Materials Testing ,medicine ,Articular cartilage repair ,Animals ,Orthopedics and Sports Medicine ,Cells, Cultured ,Analysis of Variance ,Tissue Engineering ,Tissue Scaffolds ,business.industry ,Cartilage ,Anatomy ,In vitro ,Biomechanical Phenomena ,Cell biology ,Blood ,medicine.anatomical_structure ,Membrane ,Engineered cartilage ,Surgery ,business - Abstract
The aim of this study was to investigate the in vitro effect of different concentrations of blood on the morphological and biochemical properties of engineered cartilage. Previous studies have demonstrated a negative effect of blood on native cartilage; however, the effect of the contact of blood on engineered cartilage is unclear. Articular chondrocytes were isolated from swine joints, expanded in monolayer culture, and seeded onto collagen membranes. The seeded membranes were cultured for 3 days in the presence of different concentrations of peripheral blood. Some samples were retrieved at the end of the blood contact, others after 21 additional days of standard culture conditions, in order to investigate the “long-term effect” of the blood contact. All seeded samples showed an increase in the weight and an evident cartilage-like matrix production. A concentration-dependent reduction in the mitochondrial activity due to blood contact was shown at the earlier culture time, followed by a partial recover at the longer culture time. A blood contact of 3 days affected the chondrocytes’ activity and determined a delay in the maturation of the engineered cartilage. These findings have clinical relevance, as autologous chondrocytes seeded onto biological scaffolds has become an established surgical method for articular cartilage repair. Therefore, further investigation into material sciences should be encouraged for the development of scaffold protecting the reparative cells from the blood insult.
- Published
- 2010
- Full Text
- View/download PDF
30. Healing of meniscal tissue by cellular fibrin glue: an in vivo study
- Author
-
Laura Mangiavini, F. Vitari, A. Pozzi, Federica Boschetti, Cinzia Domeneghini, Giuseppe M. Peretti, Celeste Scotti, and Gianfranco Fraschini
- Subjects
Scaffold ,Sus scrofa ,Mice, Nude ,Fibrin Tissue Adhesive ,Meniscus (anatomy) ,Matrix (biology) ,Menisci, Tibial ,Fibrin ,Mice ,Chondrocytes ,Tissue engineering ,medicine ,Animals ,Orthopedics and Sports Medicine ,Fibrin glue ,Wound Healing ,Tissue Engineering ,biology ,business.industry ,Anatomy ,Tibial Meniscus Injuries ,medicine.anatomical_structure ,biology.protein ,Tissue Adhesives ,Surgery ,business ,Wound healing ,Biomedical engineering - Abstract
Menisci represent fundamental structures for the maintenance of knee homeostasis, playing a key role in knee biomechanics. However, their intrinsic regenerative potential is poor. As a consequence, when a lesion occurs and the meniscus is partially removed by surgery, knee mechanics is subject to dramatic changes. These have been demonstrated to lead often to the development of early osteoarthritis. Therefore, menisci should be repaired whenever possible. In the last decades, tissue engineering approaches have been advocated to improve the reparative processes of joint tissues. In this study, the bonding capacity of an articular chondrocytes-fibrin glue hydrogel was tested as a biologic glue to improve the bonding between two swine meniscal slices in a nude mouse model. The composites were wrapped with acellular fibrin glue and implanted in subcutaneous pouches of nude mice for 4 weeks. Upon retrieval, a firm gross bonding was observed in the experimental samples while none of the control samples, prepared with acellular fibrin glue at the interface, presented any sign of bonding. This was consistent with the histological and scanning electron microscope findings. In particular, a fibrocartilaginous tissue was found at the interface between the meniscal slices, partially penetrating the native meniscus tissue. In order to overcome the lack of regenerative properties of the meniscus, the rationale of using cellular fibrin glue is that fibrin provides immediate stability while carrying cells in the site of lesion. Moreover, fibrin gel is recognized as an optimal scaffold for cell embedding and for promoting fibrocartilaginous differentiation of the cells which synthesize matrix having healing property. These results demonstrated the potential of this model for improving the meniscal bonding. However, further orthotopic studies in a large animal model are needed to evaluate its potential for clinical application.
- Published
- 2009
- Full Text
- View/download PDF
31. Bonding of meniscal tissue: a nude mouse repair model
- Author
-
Giuseppe M. Peretti, Celeste Scotti, F. Vitari, Laura Mangiavini, Cinzia Domeneghini, Gianfranco Fraschini, and A. Pozzi
- Subjects
biology ,Meniscal tissue ,business.industry ,Knee biomechanics ,Anatomy ,musculoskeletal system ,biology.organism_classification ,Nude mouse ,Tissue engineering ,Medicine ,Orthopedics and Sports Medicine ,business ,Fibrin glue ,Meniscus repair ,Early osteoarthritis ,Biomedical engineering ,Large animal - Abstract
Meniscus repair is a current clinical challenge. Menisci play a fundamental role in knee biomechanics, but they lack intrinsic regenerative properties. Conse quently, when a tear occurs and the meniscus is removed surgically, even partially, crucial changes in knee homeostasis take place, often leading to the development of early osteoarthritis. In recent decades tissue engineering approaches have been advocated to improve the reparative processes of joint tissues. In this study, the bonding capacity of isolated chondrocytes was analysed in a nude mouse meniscus repair model: a swine chondrocyte-fibrin glue suspension was utilised as a biologic glue to improve bonding between two meniscal slices obtained from swine menisci. The composites were wrapped with acellular fibrin glue and implanted in subcutaneous pouches of nude mice for four weeks. Upon retrieval, a firm gross bonding was observed. This was consistent with the histological findings. In particular, a fibrocartilaginous tissue was found at the interface between the meniscal slices, having some penetration buds arising from the neo-tissue. These results demonstrated the potential of this model for improving meniscal bonding. However, further orthotopic studies in a large animal model are needed to evaluate its feasibility in clinical practice.
- Published
- 2008
- Full Text
- View/download PDF
32. Extracorporeal shock wave treatment of humeral nonunion: a case report
- Author
-
Pietro Ciampi, Giuseppe M. Peretti, Celeste Scotti, and Gianfranco Fraschini
- Subjects
medicine.medical_specialty ,Sling (implant) ,Sports medicine ,medicine.diagnostic_test ,business.industry ,Radiography ,Nonunion ,Physical examination ,musculoskeletal system ,medicine.disease ,Extracorporeal shock wave ,Surgery ,medicine.anatomical_structure ,medicine ,Outpatient clinic ,Orthopedics and Sports Medicine ,Humerus ,business - Abstract
Fracture of the humerus represents a common problem among the young and elderly populations. Although humerus fractures usually heal uneventfully, nonunions can sometimes occur. We present a case of humeral nonunion managed with shock wave therapy in the outpatient setting. A 62-year-old woman with a closed comminuted fracture of the proximal third of the humerus came to our attention 6 months after the trauma with a hypertrophic nonunion. Radiographs showed a hypertrophic callus with a fracture gap of 4 mm. We performed ten shock wave treatments in the outpatient department, with an interval of 60 days between each single treatment. Each session consisted of 2000 impulses at 0.86 mJ/mm2 applied in two planes. No anesthesia was given during the treatment. The patient received a sling to support the treated arm during the first days following each treatment. Follow-up assessment by radiographic and physical examination, performed after 4 weeks and at 3, 6, 12, 18 and 24 months, showed complete bony union and cortical bridging, achieved at the end of the treatment. We believe that this method is a safe and effective alternative to surgery for the treatment of chronic hypertrophic nonunions. Moreover, in case such treatment is unsuccessful, subsequent surgery is not precluded.
- Published
- 2007
- Full Text
- View/download PDF
33. Fresh osteochondral allografts in the knee: only a salvage procedure?
- Author
-
Alberto, Gobbi, Celeste, Scotti, John G, Lane, and Giuseppe M, Peretti
- Subjects
Editorial ,eye diseases - Abstract
The role of fresh allogeneic osteochondral allograft transplantation (OCA) in the cartilage repair algorithm has been long debated and this procedure is primarily considered as a salvage procedure, to be used when other, simple, techniques have failed. Gracitelli et al. in a retrospective comparison of patients who received OCA as primary treatment or as a salvage procedure, demonstrates that the outcome of this procedure is minimally influenced by a previous failed treatment and that OCA represents an effective solution for both primary and revision surgery of chondral and osteochondral lesions of the knee. In particular, optimal indications for OCA seem to be revision of previously failed bone marrow stimulation techniques with an impaired subchondral bone plate and primary treatment of large osteochondral defects.
- Published
- 2015
34. Erratum to: One-step surgery with multipotent stem cells and Hyaluronan-based scaffold for the treatment of full-thickness chondral defects of the knee in patients older than 45 years
- Author
-
Georgios Karnatzikos, Giuseppe M. Peretti, Celeste Scotti, Abhishek Mudhigere, Marc Castro, and Alberto Gobbi
- Subjects
Adult ,Cartilage, Articular ,Male ,medicine.medical_specialty ,Knee Joint ,Population ,Mesenchymal Stem Cell Transplantation ,Transplantation, Autologous ,Young Adult ,medicine ,Humans ,Orthopedics and Sports Medicine ,Prospective Studies ,Hyaluronic Acid ,Prospective cohort study ,education ,Stem cell transplantation for articular cartilage repair ,education.field_of_study ,Tissue Scaffolds ,Viscosupplements ,business.industry ,Cartilage ,Mesenchymal stem cell ,Age Factors ,Middle Aged ,Magnetic Resonance Imaging ,Surgery ,medicine.anatomical_structure ,Multipotent Stem Cell ,Orthopedic surgery ,Female ,Erratum ,business ,Cartilage Diseases - Abstract
The aim of this study is to prospectively evaluate the medium-term effectiveness and regenerative capability of autologous adult mesenchymal stem cells, harvested as bone marrow aspirate concentrate (BMAC), along with a hyaluronan-based scaffold (Hyalofast) in the treatment of ICRS grade 4 chondral lesions of the knee joint, in patients older than 45 years.A study group of 20 patients with an age45 years (mean 50.0 ± 4.1 years) was compared to a control group of 20 patients with an age45 years (mean 36.6 ± 5.0). Patients were prospectively evaluated for 4 years. All patients were evaluated with MRI, KOOS, IKDC, VAS and Tegner scores preoperatively and at two-year and final follow-up.At final follow-up, all scores significantly improved (P 0.001) as follows: all KOOS score categories; Tegner 2 (range 0-4) to 6 (range 4-8) and 3 (range 0-6) to 6 (range 3-10); IKDC subjective (39.2 ± 16.5 to 82.2 ± 8.9) and (40.8 ± 13.9 to 79.4 ± 14.6), in the study and control group respectively. In addition, we show that results are affected by lesion size and number but not from concomitant surgical procedures. MRI showed complete filling in 80 % of patients in the study group and 71 % of patients in the control group. Histological analysis conducted in three patients from the study and two patients from the control group revealed good tissue repair with a variable amount of hyaline-like tissue.Treatment of cartilage lesions with BMAC and Hyalofast is a viable and effective option that is mainly affected by lesion size and number and not by age. In particular, it allows to address the45 years population with functional outcomes that are comparable to younger patients at final follow-up.Prospective cohort study, Level II.
- Published
- 2017
- Full Text
- View/download PDF
35. Generation and characterization of osteochondral grafts with human nasal chondrocytes
- Author
-
Marina, Barandun, Lukas Daniel, Iselin, Francesco, Santini, Michele, Pansini, Celeste, Scotti, Daniel, Baumhoer, Oliver, Bieri, Ueli, Studler, Dieter, Wirz, Martin, Haug, Marcel, Jakob, Dirk Johannes, Schaefer, Ivan, Martin, and Andrea, Barbero
- Subjects
Cartilage, Articular ,Male ,Chondrocytes ,Tissue Engineering ,Interleukin-1beta ,Humans ,Female ,Middle Aged ,Nose ,Magnetic Resonance Imaging ,Aged ,Glycosaminoglycans - Abstract
We investigated whether nasal chondrocytes (NC) can be used to generate composite constructs with properties necessary for the repair of osteochondral (OC) lesions, namely maturation, integration and capacity to recover from inflammatory burst. OC grafts were fabricated by combining engineered cartilage tissues (generated by culturing NC or articular chondrocytes - AC - onto Chondro-Gide® matrices) with devitalized spongiosa cylinders (Tutobone®). OC tissues were then exposed to IL-1β for three days and cultured for additional 2 weeks in the absence of IL-1β. Cartilage maturation extent was assessed (immune) histologically, biochemically and by delayed gadolinium-enhanced magnetic resonance imaging of cartilage (dGEMRIC) while cartilage/bone integration was assessed using a peel-off mechanical test. The use of NC as compared to AC allowed for more efficient cartilage matrix accumulation and superior integration of the cartilage/bone layers. dGEMRIC and biochemical analyzes of the OC constructs showed a reduced glycosaminoglycan (GAG) contents upon IL-1β administration. Cartilaginous matrix contents and integration forces returned to baseline up on withdrawal of IL-1β. By having a cartilage layer well developed and strongly integrated to the subchondral layer, OC tissues generated with NC may successfully engraft in an inflammatory post-surgery joint environment.
- Published
- 2014
36. Adult human neural crest-derived cells for articular cartilage repair
- Author
-
Brigitte von Rechenberg, Marcel Jakob, Benjamin E. Pippenger, Marcus Mumme, Ivan Martin, Thomas Schwamborn, Pierre Mainil-Varlet, Alfredo Procino, Sandra Feliciano, Andrea Barbero, Celeste Scotti, Clemente Cillo, Karoliina Pelttari, Pelttari, K, Pippenger, B, Mumme, M, Feliciano, S, Scotti, C, Mainil Varlet, P, Procino, A, von Rechenberg, B, Schwamborn, T, Jakob, M, Cillo, Clemente, Barbero, A, and Martin, I.
- Subjects
Adult ,Cartilage, Articular ,animal structures ,Knee Joint ,Pilot Projects ,Biology ,Transplantation, Autologous ,Mice ,medicine ,Articular cartilage repair ,Animals ,Humans ,Stem cell transplantation for articular cartilage repair ,Cell Proliferation ,Homeodomain Proteins ,Wound Healing ,Neuronal Plasticity ,Neuroectoderm ,Hyaline cartilage ,Cartilage ,Gene Expression Profiling ,Goats ,Mesenchymal stem cell ,Neural crest ,General Medicine ,Anatomy ,Middle Aged ,Coculture Techniques ,Cell biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Neural Crest ,embryonic structures ,Stem cell - Abstract
In embryonic models and stem cell systems, mesenchymal cells derived from the neuroectoderm can be dis- tinguished from mesoderm-derived cells by their Hox-negative profile—a phenotype associated with enhanced capacity of tissue regeneration. We investigated whether developmental origin and Hox negativity correlated with self-renewal and environmental plasticity also in differentiated cells from adults. Using hyaline cartilage as a model, we showed that adult human neuroectoderm-derived nasal chondrocytes (NCs) can be constitutively distinguished from mesoderm-derived articular chondrocytes (ACs) by lack of expression of specific HOX genes, including HOXC4 and HOXD8. In contrast to ACs, serially cloned NCs could be continuously reverted from differen- tiated to dedifferentiated states, conserving the ability to form cartilage tissue in vitro and in vivo. NCs could also be reprogrammed to stably express Hox genes typical of ACs upon implantation into goat articular cartilage defects, directly contributing to cartilage repair. Our findings identify previously unrecognized regenerative properties of HOX-negative differentiated neuroectoderm cells in adults, implying a role for NCs in the unmet clinical challenge of articular cartilage repair. An ongoing phase 1 clinical trial preliminarily indicated the safety and feasibility of autol- ogous NC-based engineered tissues for the treatment of traumatic articular cartilage lesions.
- Published
- 2014
37. CORR Insights®: Mesenchymal Stem Cells in Synovial Fluid Increase After Meniscus Injury
- Author
-
Celeste Scotti
- Subjects
Adult ,medicine.medical_specialty ,Time Factors ,Adolescent ,Knee Injuries ,Meniscus (anatomy) ,Menisci, Tibial ,CORR Insights ,Epitopes ,Young Adult ,Synovial Fluid ,Medicine ,Synovial fluid ,Humans ,Orthopedics and Sports Medicine ,Cell Lineage ,Child ,Cells, Cultured ,Cell Proliferation ,Wound Healing ,business.industry ,Mesenchymal stem cell ,Conflict of interest ,Mesenchymal Stem Cells ,General Medicine ,Middle Aged ,Surgery ,Tibial Meniscus Injuries ,Up-Regulation ,medicine.anatomical_structure ,Family medicine ,Case-Control Studies ,Related research ,business ,Knee injuries ,Biomarkers - Abstract
Although relatively uncommon, spontaneous healing from a meniscus injury has been observed even within the avascular area. This may be the result of the existence of mesenchymal stem cells in synovial fluid.The purpose of this study was to investigate whether mesenchymal stem cells existed in the synovial fluid of the knee after meniscus injury.Synovial fluid was obtained from the knees of 22 patients with meniscus injury just before meniscus surgery and from 8 volunteers who had no history of knee injury. The cellular fraction of the synovial fluid was cultured for 14 days followed by analysis for multilineage potential and presentation of surface antigens characteristic of mesenchymal stem cells. Colony-forming efficiency and proliferation potential were also compared between the two groups.Cells with characteristics of mesenchymal stem cells were observed in the synovial fluid of injured knees to a much greater degree than in uninjured knees. The colony-forming cells derived from the synovial fluid of the knee with meniscus injury had multipotentiality and surface epitopes identical to mesenchymal stem cells. The average number of colony formation, obtained from 1 mL of synovial fluid, in meniscus-injured knees was 250, higher than that from healthy volunteers, which was 0.5 (p 0.001). Total colony number per synovial fluid volume was positively correlated with the postinjury period (r = 0.77, p 0.001).Mesenchymal stem cells were found to exist in synovial fluid from knees after meniscus injury. Mesenchymal stem cells were present in higher numbers in synovial fluid with meniscus injury than in normal knees. Total colony number per synovial fluid volume was positively correlated with the postinjury period.Our current human study and previous animal studies suggest the possibility that mesenchymal stem cells in synovial fluid increase after meniscus injury contributing to spontaneous meniscus healing.
- Published
- 2014
38. Engineering of a functional bone organ through endochondral ossification
- Author
-
Andrea Barbero, Atanas Todorov, Markus G. Manz, Adam Papadimitropoulos, Elia Piccinini, Hitoshi Takizawa, Celeste Scotti, Ivan Martin, Paul Bourgine, University of Zurich, and Martin, I
- Subjects
Adult ,Mature Bone ,Interleukin-1beta ,Transplantation, Heterologous ,Mice, Nude ,Neovascularization, Physiologic ,610 Medicine & health ,02 engineering and technology ,Biology ,Mesenchymal Stem Cell Transplantation ,Regenerative Medicine ,Models, Biological ,03 medical and health sciences ,Mice ,Bone Marrow ,Osteogenesis ,Bone cell ,medicine ,Bone organ ,Animals ,Humans ,Stem Cell Niche ,Endochondral ossification ,030304 developmental biology ,Bone morphogenesis ,Bone Marrow Transplantation ,0303 health sciences ,1000 Multidisciplinary ,Multidisciplinary ,Tissue Engineering ,Tissue Scaffolds ,Hematopoietic stem cell ,Mesenchymal Stem Cells ,Anatomy ,Biological Sciences ,021001 nanoscience & nanotechnology ,Hematopoietic Stem Cells ,Cell biology ,Hematopoiesis ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Cartilage ,Intramembranous ossification ,10032 Clinic for Oncology and Hematology ,Bone marrow ,0210 nano-technology - Abstract
Embryonic development, lengthening, and repair of most bones proceed by endochondral ossification, namely through formation of a cartilage intermediate. It was previously demonstrated that adult human bone marrow-derived mesenchymal stem/stromal cells (hMSCs) can execute an endochondral program and ectopically generate mature bone. Here we hypothesized that hMSCs pushed through endochondral ossification can engineer a scaled-up ossicle with features of a “bone organ,” including physiologically remodeled bone, mature vasculature, and a fully functional hematopoietic compartment. Engineered hypertrophic cartilage required IL-1β to be efficiently remodeled into bone and bone marrow upon subcutaneous implantation. This model allowed distinguishing, by analogy with bone development and repair, an outer, cortical-like perichondral bone, generated mainly by host cells and laid over a premineralized area, and an inner, trabecular-like, endochondral bone, generated mainly by the human cells and formed over the cartilaginous template. Hypertrophic cartilage remodeling was paralleled by ingrowth of blood vessels, displaying sinusoid-like structures and stabilized by pericytic cells. Marrow cavities of the ossicles contained phenotypically defined hematopoietic stem cells and progenitor cells at similar frequencies as native bones, and marrow from ossicles reconstituted multilineage long-term hematopoiesis in lethally irradiated mice. This study, by invoking a “developmental engineering” paradigm, reports the generation by appropriately instructed hMSC of an ectopic “bone organ” with a size, structure, and functionality comparable to native bones. The work thus provides a model useful for fundamental and translational studies of bone morphogenesis and regeneration, as well as for the controlled manipulation of hematopoietic stem cell niches in physiology and pathology.
- Published
- 2013
39. Animal models for meniscus repair and regeneration
- Author
-
Daniela, Deponti, Alessia, Di Giancamillo, Celeste, Scotti, Giuseppe M, Peretti, and Ivan, Martin
- Subjects
Wound Healing ,Sheep ,Tissue Engineering ,Cell- and Tissue-Based Therapy ,Biocompatible Materials ,Regenerative Medicine ,Menisci, Tibial ,Dogs ,Models, Animal ,Animals ,Humans ,Regeneration ,Knee ,Rabbits - Abstract
The meniscus plays an important role in knee function and mechanics. Meniscal lesions, however, are common phenomena and this tissue is not able to achieve spontaneous successful repair, particularly in the inner avascular zone. Several animal models have been studied and proposed for testing different reparative approaches, as well as for studying regenerative methods aiming to restore the original shape and function of this structure. This review summarizes the gross anatomy, function, ultrastructure and biochemical composition of the knee meniscus in several animal models in comparison with the human meniscus. The relevance of the models is discussed from the point of view of basic research as well as of clinical translation for meniscal repair, substitution and regeneration. Finally, the advantages and disadvantages of each model for various research directions are critically discussed.
- Published
- 2012
40. Response of human engineered cartilage based on articular or nasal chondrocytes to interleukin-1? and low oxygen
- Author
-
Andrea Osmokrovic, Ivan Martin, Sylvie Miot, Andrea Barbero, Giuseppe M. Peretti, Celeste Scotti, and Francine Wolf
- Subjects
Adult ,Cartilage, Articular ,Male ,Interleukin-1beta ,Biomedical Engineering ,Bioengineering ,Matrix metalloproteinase ,Cartilage Oligomeric Matrix Protein ,Nose ,Biochemistry ,Biomaterials ,Glycosaminoglycan ,Andrology ,Extracellular matrix ,Epitopes ,Chondrocytes ,Tissue engineering ,medicine ,Humans ,Matrilin Proteins ,Collagen Type II ,Cells, Cultured ,Aged ,Glycoproteins ,Glycosaminoglycans ,Cartilage oligomeric matrix protein ,Aged, 80 and over ,Extracellular Matrix Proteins ,biology ,Tissue Engineering ,Tissue Scaffolds ,Chemistry ,Cartilage ,Interleukin ,Original Articles ,Middle Aged ,Chondrogenesis ,Matrix Metalloproteinases ,Oxygen ,medicine.anatomical_structure ,Immunology ,biology.protein ,Female - Abstract
Previous studies showed that human nasal chondrocytes (HNC) exhibit higher proliferation and chondrogenic capacity as compared to human articular chondrocytes (HAC). To consider HNC as a relevant alternative cell source for the repair of articular cartilage defects it is necessary to test how these cells react when exposed to environmental factors typical of an injured joint. We thus aimed this study at investigating the responses of HNC and HAC to exposure to interleukin (IL)-1? and low oxygen. For this purpose HAC and HNC harvested from the same donors (N=5) were expanded in vitro and then cultured in pellets or collagen-based scaffolds at standard (19%) or low oxygen (5%) conditions. Resulting tissues were analyzed after a short (3 days) exposure to IL-1?, mimicking the initially inflammatory implantation site, or following a recovery time (1 or 2 weeks for pellets and scaffolds, respectively). After IL-1? treatment, constructs generated by both HAC and HNC displayed a transient loss of GAG (up to 21.8% and 36.8%, respectively) and, consistently, an increased production of metalloproteases (MMP)-1 and -13. Collagen type II and the cryptic fragment of aggrecan (DIPEN), both evaluated immunohistochemically, displayed a trend consistent with GAG and MMPs production. HNC-based constructs exhibited a more efficient recovery upon IL-1? withdrawal, resulting in a higher accumulation of GAG (up to 2.6-fold) compared to the corresponding HAC-based tissues. On the other hand, HAC displayed a positive response to low oxygen culture, while HNC were only slightly affected by oxygen percentage. Collectively, under the conditions tested mimicking the postsurgery articular environment, HNC retained a tissue-forming capacity, similar or even better than HAC. These results represent a step forward in validating HNC as a cell source for cartilage tissue engineering strategies.
- Published
- 2012
- Full Text
- View/download PDF
41. Perspective on the evolution of cell-based bone tissue engineering strategies
- Author
-
Ivan Martin, Michael Heberer, Simone Schreiner, Marcel Jakob, Franziska Saxer, Patrick Studer, Arnaud Scherberich, and Celeste Scotti
- Subjects
medicine.medical_specialty ,Bone Regeneration ,Bone Transplantation ,Bone development ,Tissue Engineering ,Computer science ,Clinical performance ,Bone tissue engineering ,Surgery ,Clinical Practice ,Bioreactors ,Risk analysis (engineering) ,Tissue engineering ,medicine ,Animals ,Humans ,Bone regeneration ,Cell based - Abstract
Despite the compelling clinical needs in enhancing bone regeneration and the potential offered by the field of tissue engineering, the adoption of cell-based bone graft substitutes in clinical practice is limited to date. In fact, no study has yet convincingly demonstrated reproducible clinical performance of tissue-engineered implants and at least equivalent cost-effectiveness compared to the current treatment standards. Here, we propose and discuss how tissue engineering strategies could be evolved towards more efficient solutions, depicting three different experimental paradigms: (i) bioreactor-based production; (ii) intraoperative manufacturing, and (iii) developmental engineering. The described approaches reflect the need to streamline graft manufacturing processes while maintaining the potency of osteoprogenitors and recapitulating the sequence of biological steps occurring during bone development, including vascularization. The need to combine the assessment of efficacy of the different strategies with the understanding of their mechanisms of action in the target regenerative processes is highlighted. This will be crucial to identify the necessary and sufficient set of signals that need to be delivered at the injury or defect site and should thus form the basis to define release criteria for reproducibly effective engineered bone graft substitutes.
- Published
- 2012
42. Surgical treatment of thoracic outlet syndrome in young adults: single centre experience with minimum three-year follow-up
- Author
-
Giuseppe M. Peretti, Roberto Chiesa, Gianfranco Fraschini, Celeste Scotti, Simonetta Gerevini, Pietro Ciampi, Francesco De Cobelli, Ciampi, P, Scotti, C, Gerevini, S, DE COBELLI, Francesco, Chiesa, Roberto, Fraschini, G, and Peretti G., M.
- Subjects
Thoracic outlet ,Adult ,Male ,medicine.medical_specialty ,Decompression ,First rib resection ,Young Adult ,Postoperative Complications ,Surveys and Questionnaires ,medicine ,Humans ,Minimally Invasive Surgical Procedures ,Orthopedics and Sports Medicine ,Thoracic outlet syndrome ,Retrospective Studies ,Original Paper ,business.industry ,Middle Aged ,medicine.disease ,Decompression, Surgical ,Clavicle ,Magnetic Resonance Imaging ,Surgery ,Radiography ,Venous thrombosis ,Thoracic Outlet Syndrome ,medicine.anatomical_structure ,Treatment Outcome ,Patient Satisfaction ,Orthopedic surgery ,Cervical Vertebrae ,Female ,business ,Cervical vertebrae ,Follow-Up Studies - Abstract
"Thoracic outlet syndrome is an often misdiagnosed syndrome which consists of a neurovascular compression at the upper thoracic outlet. The clinical presentation can be variable, ranging from mild symptoms to venous thrombosis and muscle atrophy. Many aetiologies, both congenital and acquired, related either to bony or soft tissue anomalies, have been associated with this syndrome. As a consequence, the diagnosis is often challenging and sometimes it can be obtained only with surgical exploration. Additionally, no specific clinical test is considered diagnostic of thoracic outlet syndrome. However, the recent advances in imaging techniques together with a careful clinical evaluation give the surgeon the chance to recognize the constricting anatomy before surgery in many cases. No standard surgical procedure has been identified; however, in literature the largest series have been treated with transaxillary first rib resection. Here we report our experience in the surgical treatment of this syndrome with a minimum follow-up of three years. Our approach consists of performing a supraclavicular decompression without routine first rib resection. This allows for identifying and removing the constricting anatomy in most cases, with satisfactory results in 96.9% of patients and a low complication rate."
- Published
- 2010
43. Engineering human cell-based, functionally integrated osteochondral grafts by biological bonding of engineered cartilage tissues to bony scaffolds
- Author
-
Dirk J. Schaefer, Victor Valderrabano, Francine Wolf, Andrea Barbero, Christian Candrian, Vivienne Bürgin, Dieter Wirz, Ivan Martin, Celeste Scotti, Alma U. Daniels, and Marcel Jakob
- Subjects
Adult ,Cartilage, Articular ,Male ,Bone sialoprotein ,Scaffold ,Materials science ,Cell Culture Techniques ,Biophysics ,Transplants ,Bioengineering ,Bone and Bones ,Fibrin ,Cartilage tissue engineering ,Chondrocyte ,Biomaterials ,Extracellular matrix ,Osseointegration ,Materials Testing ,medicine ,Humans ,Collagen Type II ,Cells, Cultured ,Aged ,Aged, 80 and over ,Tissue Engineering ,Tissue Scaffolds ,biology ,Reproducibility of Results ,Middle Aged ,Human cell ,Coculture Techniques ,medicine.anatomical_structure ,Mechanics of Materials ,Engineered cartilage ,Ceramics and Composites ,biology.protein ,Female ,Biomedical engineering - Abstract
In this study, we aimed at developing and validating a technique for the engineering of osteochondral grafts based on the biological bonding of a chondral layer with a bony scaffold by cell-laid extracellular matrix. Osteochondral composites were generated by combining collagen-based matrices (Chondro-Gide) containing human chondrocytes with devitalized spongiosa cylinders (Tutobone) using a fibrin gel (Tisseel). We demonstrate that separate pre-culture of the chondral layer for 3 days prior to the generation of the composite allows for (i) more efficient cartilaginous matrix accumulation than no pre-culture, as assessed histologically and biochemically, and (ii) superior biological bonding to the bony scaffold than 14 days of pre-culture, as assessed using a peel-off mechanical test, developed to measure integration of bilayered materials. The presence of the bony scaffold induced an upregulation in the infiltrated cells of the osteoblast-related gene bone sialoprotein, indicative of the establishment of a gradient of cell phenotypes, but did not affect per se the quality of the cartilaginous matrix in the chondral layer. The described strategy to generate osteochondral plugs is simple to be implemented and--since it is based on clinically compliant cells and materials--is amenable to be readily tested in the clinic.
- Published
- 2010
- Full Text
- View/download PDF
44. Effect of in vitro culture on a chondrocyte-fibrin glue hydrogel for cartilage repair
- Author
-
Giuseppe M. Peretti, Gianfranco Fraschini, Federica Boschetti, Laura Mangiavini, F. Vitari, Celeste Scotti, and Cinzia Domeneghini
- Subjects
Cartilage, Articular ,Swine ,Fibrin Tissue Adhesive ,Sensitivity and Specificity ,Chondrocyte ,Hydrogel, Polyethylene Glycol Dimethacrylate ,Statistics, Nonparametric ,Chondrocytes ,Tissue engineering ,medicine ,Articular cartilage repair ,In Situ Nick-End Labeling ,Animals ,Orthopedics and Sports Medicine ,Fibrin glue ,Cells, Cultured ,Tissue Engineering ,Tissue Scaffolds ,business.industry ,Cartilage ,Regeneration (biology) ,Anatomy ,Prostheses and Implants ,Biomechanical Phenomena ,Disease Models, Animal ,medicine.anatomical_structure ,Self-healing hydrogels ,Surgery ,business ,Biomedical engineering - Abstract
Research in tissue engineering has been focused on articular cartilage repair for more than a decade. Some pioneristic studies involved the use of hydrogels such as alginate and fibrin glue which still possess valuable potential for cartilage regeneration. One of the main issues in cartilage tissue engineering is represented by the ideal maturation of the construct, before in vivo implantation, in order to optimize matrix quality and integration. The present study was focused on the effect of in vitro culture on a fibrin glue hydrogel embedding swine chondrocytes. We performed an evaluation of the immunohistochemical and biochemical composition and of the biomechanical properties of the construct after 1 and 5 weeks of culture. We noticed that chondrocytes survived in the fibrin glue gel and enhanced their synthetic activity. In fact, DNA content remained stable, while all indices of cartilage matrix production increased (GAGs content, immunohistochemistry for collagen II and safranin-o staining). On the other hand, the biomechanical properties remained steady, indicating a gradual substitution of the hydrogel scaffold by cartilaginous matrix. This demonstrates that an optimal preculture could provide the surgeon with a better engineered cartilage for implantation. However, whether this more mature tissue will result in a more efficient regeneration of the articular surface still has to be evaluated in future investigations.
- Published
- 2009
45. A tissue engineered osteochondral plug: an in vitro morphological evaluation
- Author
-
M. Buragas, Cinzia Domeneghini, Celeste Scotti, Gianfranco Fraschini, Laura Mangiavini, C. Sosio, A. Di Giancamillo, and Giuseppe M. Peretti
- Subjects
Calcium Phosphates ,Cartilage, Articular ,Scaffold ,Swine ,Cell Culture Techniques ,Fibrin Tissue Adhesive ,Articular cartilage ,Biocompatible Materials ,Matrix (biology) ,Chondrocytes ,Tissue engineering ,Medicine ,Animals ,Orthopedics and Sports Medicine ,Fibrin glue ,Glycosaminoglycans ,Tissue Engineering ,Tissue Scaffolds ,business.industry ,Cartilage ,Anatomy ,In vitro ,medicine.anatomical_structure ,Surgery ,business ,Chondrogenesis ,Biomedical engineering - Abstract
Articular cartilage lesions have a poor intrinsic healing potential. The repair tissue is often fibrous, having insufficient biomechanical properties, which could frequently lead to the development of early osteoarthritis. In the last decade, tissue engineering approaches addressed this topic in order to restore joint function with a differentiated and functional tissue. Many biomaterials and techniques have been proposed and some of them applied in clinical practice, even though several concerns have been raised on the quality of the engineered tissue and on its integration in the host joint. In this study, we focused on engineering in vitro a biphasic composite made of cellular fibrin glue and a calcium-phosphate scaffold. Biphasic composites are the latest products of tissue engineering applied to articular cartilage and they seem to allow a more efficient integration of the engineered tissue with the host. However, a firm in vitro bonding between the two components of the composite is a necessary condition to validate this model. Our study demonstrated a gross and microscopic integration of the two components and a cartilage-like quality of the newly formed matrix. Moreover, we noticed an improvement of this integration and GAGs production during the in vitro culture.
- Published
- 2007
46. Effect of blood on the morphological, biochemical and biomechanical properties of engineered cartilage
- Author
-
M. Buragas, Giuseppe M. Peretti, Federica Boschetti, Celeste Scotti, C. Bevilacqua, S. Biressi, Antonio Gigante, C. Sosio, Gianfranco Fraschini, and Laura Mangiavini
- Subjects
Cartilage, Articular ,Scaffold ,Cell Survival ,Swine ,Chondrocyte ,Chondrocytes ,Tissue engineering ,Animals ,Medicine ,Orthopedics and Sports Medicine ,Cartilage repair ,Collagen Type II ,Cells, Cultured ,Cell Proliferation ,Tissue Engineering ,Tissue Scaffolds ,business.industry ,Cartilage ,Anatomy ,Immunohistochemistry ,Peripheral blood ,In vitro ,Biomechanical Phenomena ,Mitochondria ,Blood ,medicine.anatomical_structure ,Engineered cartilage ,Surgery ,business ,Biomedical engineering - Abstract
The use of autologous chondrocytes seeded onto a biological scaffold represents a current valid tool for cartilage repair. However, the effect of the contact of blood to the engineered construct is unknown. The aim of this work was to investigate in vitro the effect of blood on the morphological, biochemical and biomechanical properties of engineered cartilage. Articular chondrocytes were enzymatically isolated from swine joints, expanded in monolayer culture and seeded onto collagen membranes for 2 weeks. Then, the seeded membranes were placed for 3 days in contact with peripheral blood, which was obtained from animals of the same species and diluted with a standard medium. As controls, some samples were left in the standard medium. After the 3 days' contact, some samples were retrieved for analysis; others were returned to standard culture conditions for 21 additional days, in order to investigate the "long-term effect" of the blood contact. Upon retrieval, all seeded samples showed increasing sizes and weights over time. However, the samples exposed to blood presented lower values with respect to the controls. Biochemical evaluation demonstrated a reduction in the mitochondrial activity due to blood contact at the early culture time (3 days post blood contact), followed by a partial recovery at the longer culture time (21 days post blood contact). Histological evaluation demonstrated evident cartilage-like matrix production for both groups. Biomechanical data showed a reduction of the values, followed by stabilization, regardless of the presence of blood. Based on the data obtained in this study, we can conclude that blood contact affects the chondrocyte activity and determines a delay in the dimensional growth of the engineered cartilage; however, at the experimental times utilized in this study, this delay did not affect the histological pattern and the biomechanical properties of the construct.
- Published
- 2007
47. Solitary psoas muscle metastasis from renal cell carcinoma
- Author
-
Celeste Scotti, Gianfranco Fraschini, Anna Borri, Francesca Fontana, and Francesco Camnasio
- Subjects
Pathology ,medicine.medical_specialty ,Text mining ,business.industry ,Renal cell carcinoma ,General surgery ,medicine ,Surgery ,General Medicine ,medicine.disease ,business ,Metastasis - Published
- 2010
- Full Text
- View/download PDF
48. Pseudoaneurysm overlying an osteochondroma: a noteworthy complication
- Author
-
Francesco Camnasio, Enrico Maria Marone, Giuseppe M. Peretti, Celeste Scotti, Roberto Chiesa, Laura Elisabetta Brasca, Alessandro Del Maschio, Gianfranco Fraschini, Scotti, C, Marone, Em, Brasca, Le, Peretti, Gm, Chiesa, Roberto, DEL MASCHIO, Alessandro, Fraschini, G, and Camnasio, F.
- Subjects
Osteochondroma ,Male ,medicine.medical_specialty ,Adolescent ,Brachial Artery ,Case Report ,Bone Neoplasms ,Pseudoaneurysm ,Aneurysm ,medicine.artery ,medicine ,Humans ,Humerus ,Orthopedics and Sports Medicine ,cardiovascular diseases ,Brachial artery ,business.industry ,Soft tissue mass ,Soft tissue ,medicine.disease ,Surgery ,medicine.anatomical_structure ,cardiovascular system ,Radiology ,Differential diagnosis ,Complication ,business ,Aneurysm, False - Abstract
Pseuodaneurysms are an extremely rare complication of osteochondromas. We describe a case of traumatic pseudoaneurysm of the brachial artery presenting as a soft tissue mass in a patient who was treated for an osteochondroma 3 years earlier. This case demonstrates that radiographic follow-up of large osteochondromas is mandatory and that, in patients with soft tissue masses and a history of osteochondroma, pseudoaneurysms should be included in the differential diagnosis.
- Full Text
- View/download PDF
49. Modular prostheses in the treatment of proximal humerus metastases: review of 40 cases
- Author
-
Giuseppe M. Peretti, Gianfranco Fraschini, Francesca Fontana, Celeste Scotti, and Francesco Camnasio
- Subjects
medicine.medical_specialty ,Bone disease ,Sports medicine ,business.industry ,Original ,Proximal humerus ,medicine.medical_treatment ,Bone metastases ,Modular prostheses ,Pathological fracture ,Cancer ,medicine.disease ,Prosthesis ,Rheumatology ,Surgery ,medicine.anatomical_structure ,Internal medicine ,Orthopedic surgery ,medicine ,Humerus ,Orthopedics and Sports Medicine ,business ,Pathological - Abstract
BackgroundThe humerus is the second most common site of metastatic bone disease involving long bones. Tumors which have a predilection for dissemination to bone are those of breast, prostate, thyroid, lung and kidney. The rationale for surgical treatment of these lesions is to prevent or treat pathological fractures in order to relieve pain and improve function.Materials and methodsForty patients who had resection of the proximal humerus for metastatic bone disease and reconstruction with a modular prosthesis were retrospectively reviewed.ResultsMean functional outcome was 73.1% (Enneking score) and better results were achieved when a reverse prosthesis was implanted. Overall survival was 70% at 1 year, 42.5% at 2 years and 20% at 5 years. Local recurrence occurred in 4 patients, each of whom had initially been treated for a pathological fracture.ConclusionsIt is important to follow rational guidelines, like those of Capanna and Mirels, in order to prevent pathological fractures and to give the patient a definitive treatment, as the advances in the management of cancer prolong the survival of these patients. In this series, satisfactory results were obtained, giving the patients an acceptable quality of life.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.