1
|
Largo R, Mediero A, Villa-Gomez C, Bermejo-Alvarez I, Herrero-Beaumont G. Aberrant anabolism hinders constructive metabolism of chondrocytes by pharmacotherapy in osteoarthritis. Bone Joint Res 2025; 14:199-207. [PMID: 40042132 PMCID: PMC11881514 DOI: 10.1302/2046-3758.143.bjr-2024-0241.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/03/2025] Open
Abstract
Osteoarthritis (OA) is a highly prevalent and disabling disease with an unmet therapeutic need. The characteristic cartilage loss and alteration of other joint structures result from a complex interaction of multiple risk factors, with mechanical overload consistently playing a central role. This overload generates an inflammatory response in the cartilage due to the activation of the innate immune response in chondrocytes, which occurs through various cellular mechanisms. Moreover, risk factors associated with obesity, being overweight, and metabolic syndrome enhance the inflammatory response both locally and systemically. OA chondrocytes, the only cells present in articular cartilage, are therefore inflamed and initiate an anabolic process in an attempt to repair the damaged tissue, which ultimately results in an aberrant and dysfunctional process. Under these circumstances, where the cartilage continues to be subjected to chronic mechanical stress, proposing a treatment that stimulates the chondrocytes' anabolic response to restore tissue structure does not appear to be a therapeutic target with a high likelihood of success. In fact, anabolic drugs proposed for the treatment of OA have yet to demonstrate efficacy. By contrast, multiple therapeutic strategies focused on pharmacologically managing the inflammatory component, both at the joint and systemic levels, have shown promise. Therefore, prioritizing the control of chronic innate pro-inflammatory pathways presents the most viable and promising therapeutic strategy for the effective management of OA. As research continues, this approach may offer the best opportunity to alleviate the burden of this incapacitating disease.
Collapse
Affiliation(s)
- Raquel Largo
- Joint and Bone Research Unit, Service of Rheumatology, IIS Fundación Jiménez Díaz UAM, Madrid, Spain
| | - Aranzazu Mediero
- Joint and Bone Research Unit, Service of Rheumatology, IIS Fundación Jiménez Díaz UAM, Madrid, Spain
| | - Cristina Villa-Gomez
- Joint and Bone Research Unit, Service of Rheumatology, IIS Fundación Jiménez Díaz UAM, Madrid, Spain
| | - Ismael Bermejo-Alvarez
- Joint and Bone Research Unit, Service of Rheumatology, IIS Fundación Jiménez Díaz UAM, Madrid, Spain
| | - Gabriel Herrero-Beaumont
- Joint and Bone Research Unit, Service of Rheumatology, IIS Fundación Jiménez Díaz UAM, Madrid, Spain
| |
Collapse
|
2
|
Reinhard J, Oláh T, Laschke MW, Goebel LKH, Schmitt G, Speicher-Mentges S, Menger MD, Cucchiarini M, Pape D, Madry H. Modulation of early osteoarthritis by tibiofemoral re-alignment in sheep. Osteoarthritis Cartilage 2024; 32:690-701. [PMID: 38442768 DOI: 10.1016/j.joca.2024.02.892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 01/30/2024] [Accepted: 02/18/2024] [Indexed: 03/07/2024]
Abstract
OBJECTIVE To investigate whether tibiofemoral alignment influences early knee osteoarthritis (OA). We hypothesized that varus overload exacerbates early degenerative osteochondral changes, and that valgus underload diminishes early OA. METHOD Normal, over- and underload were induced by altering alignment via high tibial osteotomy in adult sheep (n = 8 each). Simultaneously, OA was induced by partial medial anterior meniscectomy. At 6 weeks postoperatively, OA was examined in five individual subregions of the medial tibial plateau using Kellgren-Lawrence grading, quantification of macroscopic OA, semiquantitative histopathological OA and immunohistochemical type-II collagen, ADAMTS-5, and MMP-13 scoring, biochemical determination of DNA and proteoglycan contents, and micro-computed tomographic evaluation of the subchondral bone. RESULTS Multivariate analyses revealed that OA cartilaginous changes had a temporal priority over subchondral bone changes. Underload inhibited early cartilage degeneration in a characteristic topographic pattern (P ≥ 0.0983 vs. normal), in particular below the meniscal damage, avoided alterations of the subarticular spongiosa (P ≥ 0.162 vs. normal), and prevented the disturbance of otherwise normal osteochondral correlations. Overload induced early alterations of the subchondral bone plate microstructure towards osteopenia, including significantly decreased percent bone volume and increased bone surface-to-volume ratio (all P ≤ 0.0359 vs. normal). CONCLUSION The data provide high-resolution evidence that tibiofemoral alignment modulates early OA induced by a medial meniscus injury in adult sheep. Since underload inhibits early OA, these data also support the clinical value of strategies to reduce the load in an affected knee compartment to possibly decelerate structural OA progression.
Collapse
Affiliation(s)
- Jan Reinhard
- Center of Experimental Orthopaedics, Saarland University, 66421 Homburg, Germany.
| | - Tamás Oláh
- Center of Experimental Orthopaedics, Saarland University, 66421 Homburg, Germany; Cartilage Net of the Greater Region, 66421 Homburg, Germany.
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University Medical Center and Saarland University, 66421 Homburg, Germany.
| | - Lars K H Goebel
- Center of Experimental Orthopaedics, Saarland University, 66421 Homburg, Germany; Cartilage Net of the Greater Region, 66421 Homburg, Germany.
| | - Gertrud Schmitt
- Center of Experimental Orthopaedics, Saarland University, 66421 Homburg, Germany.
| | | | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University Medical Center and Saarland University, 66421 Homburg, Germany.
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University, 66421 Homburg, Germany; Cartilage Net of the Greater Region, 66421 Homburg, Germany.
| | - Dietrich Pape
- Cartilage Net of the Greater Region, 66421 Homburg, Germany; Clinique d'Eich, Centre Hospitalier de Luxembourg, Eich, 1460 Luxembourg, Germany.
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University, 66421 Homburg, Germany; Cartilage Net of the Greater Region, 66421 Homburg, Germany.
| |
Collapse
|
3
|
Cai X, Venkatesan JK, Schmitt G, Reda B, Cucchiarini M, Hannig M, Madry H. Cytotoxic effects of different mouthwash solutions on primary human articular chondrocytes and normal human articular cartilage - an in vitro study. Clin Oral Investig 2023; 27:4987-5000. [PMID: 37329464 PMCID: PMC10492729 DOI: 10.1007/s00784-023-05118-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 06/08/2023] [Indexed: 06/19/2023]
Abstract
OBJECTIVES To compare the cytotoxicity of octenidine dihydrochloride and chlorhexidine gluconate at different concentrations on primary human articular chondrocytes and cartilage. MATERIALS AND METHODS Primary cultures of human normal adult articular chondrocytes were exposed to octenidine dihydrochloride (0.001562%, 0.003125%, 0.00625%, 0.0125%, 0.025%, 0.05%, and 0.1%), chlorhexidine gluconate (0.003125%, 0.00625%, 0.0125%, 0.025%, 0.05%, 0.1%, and 0.2%), and control (Dulbecco's modified Eagle medium or phosphate-buffered saline) for 30 s. Normal human articular cartilage explants were exposed to octenidine dihydrochloride (0.1% versus control) and chlorhexidine gluconate (0.1% versus control) for 30 s. The viability of human articular chondrocytes was measured by Trypan blue staining, Cell Proliferation Reagent WST-1, and Live/Dead staining. The proliferation of human chondrocytes was measured using the Cell Proliferation Reagent WST-1. The viability of human articular cartilage explants was measured by using Live/Dead staining. RESULTS Octenidine dihydrochloride and chlorhexidine gluconate exposure decreased cell viability and proliferation in a dose-dependent manner in primary human articular chondrocytes. Octenidine dihydrochloride and chlorhexidine gluconate exposure decreased cell viability in human articular cartilage explant cultures. CONCLUSION The degree of toxicity varied between octenidine dihydrochloride and chlorhexidine gluconate, with chlorhexidine gluconate being less toxic than octenidine dihydrochloride at the same concentration. Additionally, both octenidine dihydrochloride and chlorhexidine gluconate evaluation had cytotoxic effects on human articular cartilage. Therefore, dosing for the antimicrobial mouthwash ingredients administration would ideally be determined to remain below IC50. CLINICAL RELEVANCE These data support the in vitro safety of antimicrobial mouthwashes on primary adult human articular chondrocytes.
Collapse
Affiliation(s)
- Xiaoyu Cai
- Center of Experimental Orthopaedics, Saarland University, 66421, Homburg, Germany
- Department of Spine Surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | | | - Gertrud Schmitt
- Center of Experimental Orthopaedics, Saarland University, 66421, Homburg, Germany
| | - Bashar Reda
- Clinic of Operative Dentistry, Periodontology and Preventive Dentistry, University Hospital, Saarland University, 66421, Homburg, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University, 66421, Homburg, Germany
| | - Matthias Hannig
- Clinic of Operative Dentistry, Periodontology and Preventive Dentistry, University Hospital, Saarland University, 66421, Homburg, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University, 66421, Homburg, Germany.
| |
Collapse
|
4
|
Amini M, Venkatesan JK, Nguyen TN, Liu W, Leroux A, Madry H, Migonney V, Cucchiarini M. rAAV TGF-β and FGF-2 Overexpression via pNaSS-Grafted PCL Films Stimulates the Reparative Activities of Human ACL Fibroblasts. Int J Mol Sci 2023; 24:11140. [PMID: 37446318 DOI: 10.3390/ijms241311140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/03/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Lesions in the human anterior cruciate ligament (ACL) are frequent, unsolved clinical issues due to the limited self-healing ability of the ACL and lack of treatments supporting full, durable ACL repair. Gene therapy guided through the use of biomaterials may steadily activate the processes of repair in sites of ACL injury. The goal of the present study was to test the hypothesis that functionalized poly(sodium styrene sulfonate)-grafted poly(ε-caprolactone) (pNaSS-grafted PCL) films can effectively deliver recombinant adeno-associated virus (rAAV) vectors as a means of overexpressing two reparative factors (transforming growth factor beta-TGF-β and basic fibroblast growth factor-FGF-2) in primary human ACL fibroblasts. Effective, durable rAAV reporter red fluorescent protein and candidate TGF-β and FGF-2 gene overexpression was achieved in the cells for at least 21 days, especially when pNaSS-grafted PCL films were used versus control conditions, such as ungrafted films and systems lacking vectors or films (between 1.8- and 5.2-fold differences), showing interactive regulation of growth factor production. The expression of TGF-β and FGF-2 from rAAV via PCL films safely enhanced extracellular matrix depositions of type-I/-III collagen, proteoglycans/decorin, and tenascin-C (between 1.4- and 4.5-fold differences) in the cells over time with increased levels of expression of the specific transcription factors Mohawk and scleraxis (between 1.7- and 3.7-fold differences) and without the activation of the inflammatory mediators IL-1β and TNF-α, most particularly with pNaSS-grafted PCL films relative to the controls. This work shows the value of combining rAAV gene therapy with functionalized PCL films to enhance ACL repair.
Collapse
Affiliation(s)
- Mahnaz Amini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Germany
| | - Jagadeesh K Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Germany
| | - Tuan N Nguyen
- LBPS/CSPBAT UMR CNRS 7244, Université Sorbonne Paris Nord, 93430 Villetaneuse, France
| | - Wei Liu
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Germany
| | - Amélie Leroux
- LBPS/CSPBAT UMR CNRS 7244, Université Sorbonne Paris Nord, 93430 Villetaneuse, France
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Germany
| | - Véronique Migonney
- LBPS/CSPBAT UMR CNRS 7244, Université Sorbonne Paris Nord, 93430 Villetaneuse, France
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Germany
| |
Collapse
|
5
|
Uebelhoer M, Lambert C, Grisart J, Guse K, Plutizki S, Henrotin Y. Interleukins, growth factors, and transcription factors are key targets for gene therapy in osteoarthritis: A scoping review. Front Med (Lausanne) 2023; 10:1148623. [PMID: 37077668 PMCID: PMC10106745 DOI: 10.3389/fmed.2023.1148623] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/15/2023] [Indexed: 04/05/2023] Open
Abstract
Objective Osteoarthritis (OA) is the most common degenerative joint disease, characterized by a progressive loss of cartilage associated with synovitis and subchondral bone remodeling. There is however no treatment to cure or delay the progression of OA. The objective of this manuscript was to provide a scoping review of the preclinical and clinical studies reporting the effect of gene therapies for OA. Method This review followed the JBI methodology and was reported in accordance with the PRISMA-ScR checklist. All research studies that explore in vitro, in vivo, or ex vivo gene therapies that follow a viral or non-viral gene therapy approach were considered. Only studies published in English were included in this review. There were no limitations to their date of publication, country of origin, or setting. Relevant publications were searched in Medline ALL (Ovid), Embase (Elsevier), and Scopus (Elsevier) in March 2023. Study selection and data charting were performed by two independent reviewers. Results We found a total of 29 different targets for OA gene therapy, including studies examining interleukins, growth factors and receptors, transcription factors and other key targets. Most articles were on preclinical in vitro studies (32 articles) or in vivo animal models (39 articles), while four articles were on clinical trials related to the development of TissueGene-C (TG-C). Conclusion In the absence of any DMOAD, gene therapy could be a highly promising treatment for OA, even though further development is required to bring more targets to the clinical stage.
Collapse
Affiliation(s)
| | - Cécile Lambert
- musculoSKeletal Innovative Research Lab (mSKIL), Center for Interdisciplinary Research on Medicines, University of Liège, Liège, Belgium
| | | | - Kilian Guse
- GeneQuine Biotherapeutics GmbH, Hamburg, Germany
| | | | - Yves Henrotin
- Artialis S.A., Liège, Belgium
- musculoSKeletal Innovative Research Lab (mSKIL), Center for Interdisciplinary Research on Medicines, University of Liège, Liège, Belgium
- Department of Physical Therapy and Rehabilitation, Princess Paola Hospital, Vivalia, Marche-en-Famenne, Belgium
| |
Collapse
|
6
|
Schrenker S, Cucchiarini M, Goebel L, Oláh T, Venkatesan JK, Schmitt G, Speicher-Mentges S, Maihöfer J, Gao L, Zurakowski D, Menger MD, Laschke MW, Madry H. In vivo rAAV-mediated human TGF-β overexpression reduces perifocal osteoarthritis and improves osteochondral repair in a large animal model at one year. Osteoarthritis Cartilage 2023; 31:467-481. [PMID: 36481450 DOI: 10.1016/j.joca.2022.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 09/20/2022] [Accepted: 11/03/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Osteoarthritis (OA) is a serious consequence of focal osteochondral defects. Gene transfer of human transforming growth factor beta (hTGF-β) with recombinant adeno-associated virus (rAAV) vectors offers a strategy to improve osteochondral repair. However, the long-term in vivo effects of such rAAV-mediated TGF-β overexpression including its potential benefits on OA development remain unknown. METHOD Focal osteochondral defects in minipig knees received rAAV-lacZ (control) or rAAV-hTGF-β in vivo. After one year, osteochondral repair and perifocal OA were visualized using validated macroscopic scoring, ultra-high-field MRI at 9.4 T, and micro-CT. A quantitative estimation of the cellular densities and a validated semi-quantitative scoring of histological and immunohistological parameters completed the analysis of microarchitectural parameters. RESULTS Direct rAAV-hTGF-β application induced and maintained significantly improved defect filling and safranin O staining intensity and overall cartilage repair at one year in vivo. In addition, rAAV-hTGF-β led to significantly higher chondrocyte densities within the cartilaginous repair tissue without affecting chondrocyte hypertrophy and minimized subarticular trabecular separation. Of note, rAAV-hTGF-β significantly improved the adjacent cartilage structure and chondrocyte density and reduced overall perifocal OA development after one year in vivo. CONCLUSIONS rAAV-hTGF-β treatment improves long-term osteochondral repair and delays the progression of perifocal OA in a translational model. These findings have considerable potential for targeted molecular approaches to treat focal osteochondral defects.
Collapse
Affiliation(s)
- S Schrenker
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, 66421, Homburg, Saar, Germany.
| | - M Cucchiarini
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, 66421, Homburg, Saar, Germany.
| | - L Goebel
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, 66421, Homburg, Saar, Germany.
| | - T Oláh
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, 66421, Homburg, Saar, Germany.
| | - J K Venkatesan
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, 66421, Homburg, Saar, Germany.
| | - G Schmitt
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, 66421, Homburg, Saar, Germany.
| | - S Speicher-Mentges
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, 66421, Homburg, Saar, Germany.
| | - J Maihöfer
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, 66421, Homburg, Saar, Germany.
| | - L Gao
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, 66421, Homburg, Saar, Germany.
| | - D Zurakowski
- Departments of Anesthesia and Surgery, Children's Hospital Boston, Harvard Medical School, Boston, MA, 02115, USA.
| | - M D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany.
| | - M W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany.
| | - H Madry
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, 66421, Homburg, Saar, Germany.
| |
Collapse
|
7
|
Chondrocyte Hypertrophy in Osteoarthritis: Mechanistic Studies and Models for the Identification of New Therapeutic Strategies. Cells 2022; 11:cells11244034. [PMID: 36552796 PMCID: PMC9777397 DOI: 10.3390/cells11244034] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/08/2022] [Indexed: 12/16/2022] Open
Abstract
Articular cartilage shows limited self-healing ability owing to its low cellularity and avascularity. Untreated cartilage defects display an increased propensity to degenerate, leading to osteoarthritis (OA). During OA progression, articular chondrocytes are subjected to significant alterations in gene expression and phenotype, including a shift towards a hypertrophic-like state (with the expression of collagen type X, matrix metalloproteinases-13, and alkaline phosphatase) analogous to what eventuates during endochondral ossification. Present OA management strategies focus, however, exclusively on cartilage inflammation and degradation. A better understanding of the hypertrophic chondrocyte phenotype in OA might give new insights into its pathogenesis, suggesting potential disease-modifying therapeutic approaches. Recent developments in the field of cellular/molecular biology and tissue engineering proceeded in the direction of contrasting the onset of this hypertrophic phenotype, but knowledge gaps in the cause-effect of these processes are still present. In this review we will highlight the possible advantages and drawbacks of using this approach as a therapeutic strategy while focusing on the experimental models necessary for a better understanding of the phenomenon. Specifically, we will discuss in brief the cellular signaling pathways associated with the onset of a hypertrophic phenotype in chondrocytes during the progression of OA and will analyze in depth the advantages and disadvantages of various models that have been used to mimic it. Afterwards, we will present the strategies developed and proposed to impede chondrocyte hypertrophy and cartilage matrix mineralization/calcification. Finally, we will examine the future perspectives of OA therapeutic strategies.
Collapse
|
8
|
Oláh T, Reinhard J, Laschke MW, Goebel LKH, Walter F, Schmitt G, Speicher-Mentges S, Menger MD, Cucchiarini M, Pape D, Madry H. Axial alignment is a critical regulator of knee osteoarthritis. Sci Transl Med 2022; 14:eabn0179. [PMID: 35080913 DOI: 10.1126/scitranslmed.abn0179] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Although osteoarthritis (OA), a leading cause of disability, has been associated with joint malalignment, scientific translational evidence for this link is lacking. In a clinical case study, we provide evidence of osteochondral recovery upon unloading symptomatic isolated medial tibiofemoral knee OA associated with varus malalignment. By mapping response correlations at high resolution, we identify spatially complex degenerative changes in cartilage after overloading in a clinically relevant ovine model. We further report that unloading diminishes OA cartilage degeneration and alterations of critical parameters of the subchondral bone plate in a similar topographic fashion. Last, therapeutic unloading shifted the articular cartilage and subchondral bone phenotype to normal and restored several physiological correlations disturbed in neutral and varus OA, suggesting a protective effect on the integrity of the entire osteochondral unit. Collectively, these findings identify modifiable trajectories with considerable translational potential to reduce the burden of human OA.
Collapse
Affiliation(s)
- Tamás Oláh
- Institute of Experimental Orthopaedics and Osteoarthritis Research, Saarland University, Homburg 66421, Germany.,Cartilage Net of the Greater Region, Saarland University, Homburg 66421, Germany
| | - Jan Reinhard
- Institute of Experimental Orthopaedics and Osteoarthritis Research, Saarland University, Homburg 66421, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University Medical Center and Saarland University, Homburg 66421, Germany
| | - Lars K H Goebel
- Institute of Experimental Orthopaedics and Osteoarthritis Research, Saarland University, Homburg 66421, Germany.,Cartilage Net of the Greater Region, Saarland University, Homburg 66421, Germany
| | - Frédéric Walter
- Clinique d'Eich, Centre Hospitalier de Luxembourg, Eich 1460, Luxembourg
| | - Gertrud Schmitt
- Institute of Experimental Orthopaedics and Osteoarthritis Research, Saarland University, Homburg 66421, Germany
| | - Susanne Speicher-Mentges
- Institute of Experimental Orthopaedics and Osteoarthritis Research, Saarland University, Homburg 66421, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University Medical Center and Saarland University, Homburg 66421, Germany
| | - Magali Cucchiarini
- Institute of Experimental Orthopaedics and Osteoarthritis Research, Saarland University, Homburg 66421, Germany.,Cartilage Net of the Greater Region, Saarland University, Homburg 66421, Germany
| | - Dietrich Pape
- Cartilage Net of the Greater Region, Saarland University, Homburg 66421, Germany.,Clinique d'Eich, Centre Hospitalier de Luxembourg, Eich 1460, Luxembourg
| | - Henning Madry
- Institute of Experimental Orthopaedics and Osteoarthritis Research, Saarland University, Homburg 66421, Germany.,Cartilage Net of the Greater Region, Saarland University, Homburg 66421, Germany
| |
Collapse
|
9
|
Primorac D, Molnar V, Rod E, Jeleč Ž, Čukelj F, Matišić V, Vrdoljak T, Hudetz D, Hajsok H, Borić I. Knee Osteoarthritis: A Review of Pathogenesis and State-Of-The-Art Non-Operative Therapeutic Considerations. Genes (Basel) 2020; 11:E854. [PMID: 32722615 PMCID: PMC7464436 DOI: 10.3390/genes11080854] [Citation(s) in RCA: 209] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/11/2020] [Accepted: 07/23/2020] [Indexed: 02/07/2023] Open
Abstract
Being the most common musculoskeletal progressive condition, osteoarthritis is an interesting target for research. It is estimated that the prevalence of knee osteoarthritis (OA) among adults 60 years of age or older is approximately 10% in men and 13% in women, making knee OA one of the leading causes of disability in elderly population. Today, we know that osteoarthritis is not a disease characterized by loss of cartilage due to mechanical loading only, but a condition that affects all of the tissues in the joint, causing detectable changes in tissue architecture, its metabolism and function. All of these changes are mediated by a complex and not yet fully researched interplay of proinflammatory and anti-inflammatory cytokines, chemokines, growth factors and adipokines, all of which can be measured in the serum, synovium and histological samples, potentially serving as biomarkers of disease stage and progression. Another key aspect of disease progression is the epigenome that regulates all the genetic expression through DNA methylation, histone modifications, and mRNA interference. A lot of work has been put into developing non-surgical treatment options to slow down the natural course of osteoarthritis to postpone, or maybe even replace extensive surgeries such as total knee arthroplasty. At the moment, biological treatments such as platelet-rich plasma, bone marrow mesenchymal stem cells and autologous microfragmented adipose tissue containing stromal vascular fraction are ordinarily used. Furthermore, the latter two mentioned cell-based treatment options seem to be the only methods so far that increase the quality of cartilage in osteoarthritis patients. Yet, in the future, gene therapy could potentially become an option for orthopedic patients. In the following review, we summarized all of the latest and most important research in basic sciences, pathogenesis, and non-operative treatment.
Collapse
Affiliation(s)
- Dragan Primorac
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- Eberly College of Science, The Pennsylvania State University, University Park, State College, PA 16802, USA
- The Henry C. Lee College of Criminal Justice and Forensic Sciences, University of New Haven, West Haven, CT 06516, USA
- Medical School, University of Split, 21000 Split, Croatia
- School of Medicine, Faculty of Dental Medicine and Health, University “Josip Juraj Strossmayer”, 31000 Osijek, Croatia
- School of Medicine, JJ Strossmayer University of Osijek, 31000 Osijek, Croatia
- Medical School, University of Rijeka, 51000 Rijeka, Croatia
- Medical School REGIOMED, 96 450 Coburg, Germany
- Medical School, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
| | - Vilim Molnar
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- School of Medicine, JJ Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Eduard Rod
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- School of Medicine, JJ Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Željko Jeleč
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- School of Medicine, JJ Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Nursing, University North, 48 000 Varaždin, Croatia
| | - Fabijan Čukelj
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- Medical School, University of Split, 21000 Split, Croatia
| | - Vid Matišić
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
| | - Trpimir Vrdoljak
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- Department of Orthopedics, Clinical Hospital “Sveti Duh”, 10000 Zagreb, Croatia
| | - Damir Hudetz
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- School of Medicine, JJ Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Orthopedics, Clinical Hospital “Sveti Duh”, 10000 Zagreb, Croatia
| | - Hana Hajsok
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- Medical School, University of Zagreb, 10000 Zagreb, Croatia
| | - Igor Borić
- St. Catherine Specialty Hospital, 49210 Zabok/10000 Zagreb, Croatia; (V.M.); (E.R.); (Ž.J.); (F.Č.); (V.M.); (T.V.); (D.H.); (H.H.); (I.B.)
- Medical School, University of Split, 21000 Split, Croatia
- Medical School, University of Rijeka, 51000 Rijeka, Croatia
- Medical School, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
| |
Collapse
|
10
|
Abstract
Being the most common musculoskeletal progressive condition, osteoarthritis is an interesting target for research. It is estimated that the prevalence of knee osteoarthritis (OA) among adults 60 years of age or older is approximately 10% in men and 13% in women, making knee OA one of the leading causes of disability in elderly population. Today, we know that osteoarthritis is not a disease characterized by loss of cartilage due to mechanical loading only, but a condition that affects all of the tissues in the joint, causing detectable changes in tissue architecture, its metabolism and function. All of these changes are mediated by a complex and not yet fully researched interplay of proinflammatory and anti-inflammatory cytokines, chemokines, growth factors and adipokines, all of which can be measured in the serum, synovium and histological samples, potentially serving as biomarkers of disease stage and progression. Another key aspect of disease progression is the epigenome that regulates all the genetic expression through DNA methylation, histone modifications, and mRNA interference. A lot of work has been put into developing non-surgical treatment options to slow down the natural course of osteoarthritis to postpone, or maybe even replace extensive surgeries such as total knee arthroplasty. At the moment, biological treatments such as platelet-rich plasma, bone marrow mesenchymal stem cells and autologous microfragmented adipose tissue containing stromal vascular fraction are ordinarily used. Furthermore, the latter two mentioned cell-based treatment options seem to be the only methods so far that increase the quality of cartilage in osteoarthritis patients. Yet, in the future, gene therapy could potentially become an option for orthopedic patients. In the following review, we summarized all of the latest and most important research in basic sciences, pathogenesis, and non-operative treatment.
Collapse
|
11
|
Therapeutic Delivery of rAAV sox9 via Polymeric Micelles Counteracts the Effects of Osteoarthritis-Associated Inflammatory Cytokines in Human Articular Chondrocytes. NANOMATERIALS 2020; 10:nano10061238. [PMID: 32630578 PMCID: PMC7353187 DOI: 10.3390/nano10061238] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/22/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022]
Abstract
Osteoarthritis (OA) is a prevalent joint disease linked to the irreversible degradation of key extracellular cartilage matrix (ECM) components (proteoglycans, type-II collagen) by proteolytic enzymes due to an impaired tissue homeostasis, with the critical involvement of OA-associated pro-inflammatory cytokines (interleukin 1 beta, i.e., IL-1β, and tumor necrosis factor alpha, i.e., TNF-α). Gene therapy provides effective means to re-establish such degraded ECM compounds by rejuvenating the altered OA phenotype of the articular chondrocytes, the unique cell population ubiquitous in the articular cartilage. In particular, overexpression of the highly specialized SOX9 transcription factor via recombinant adeno-associated viral (rAAV) vectors has been reported for its ability to readjust the metabolic balance in OA, in particular via controlled rAAV delivery using polymeric micelles as carriers to prevent a possible vector neutralization by antibodies present in the joints of patients. As little is known on the challenging effects of such naturally occurring OA-associated pro-inflammatory cytokines on such rAAV/polymeric gene transfer, we explored the capacity of polyethylene oxide (PEO) and polypropylene oxide (PPO)-based polymeric micelles to deliver a candidate rAAV-FLAG-hsox9 construct in human OA chondrocytes in the presence of IL-1β and TNF-α. We report that effective, micelle-guided rAAV sox9 overexpression enhanced the deposition of ECM components and the levels of cell survival, while advantageously reversing the deleterious effects afforded by the OA cytokines on these processes. These findings highlight the potentiality of polymeric micelles as effective rAAV controlled delivery systems to counterbalance the specific contribution of major OA-associated inflammatory cytokines, supporting the concept of using such systems for the treatment for chronic inflammatory diseases like OA.
Collapse
|
12
|
Alvarez-Rivera F, Rey-Rico A, Venkatesan JK, Diaz-Gomez L, Cucchiarini M, Concheiro A, Alvarez-Lorenzo C. Controlled Release of rAAV Vectors from APMA-Functionalized Contact Lenses for Corneal Gene Therapy. Pharmaceutics 2020; 12:pharmaceutics12040335. [PMID: 32283694 PMCID: PMC7238179 DOI: 10.3390/pharmaceutics12040335] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/03/2020] [Accepted: 04/07/2020] [Indexed: 12/21/2022] Open
Abstract
As an alternative to eye drops and ocular injections for gene therapy, the aim of this work was to design for the first time hydrogel contact lenses that can act as platforms for the controlled delivery of viral vectors (recombinant adeno-associated virus, rAAV) to the eye in an effective way with improved patient compliance. Hydrogels of hydroxyethyl methacrylate (HEMA) with aminopropyl methacrylamide (APMA) (H1: 40, and H2: 80 mM) or without (Hc: 0 mM) were synthesized, sterilized by steam heat (121 °C, 20 min), and then tested for gene therapy using rAAV vectors to deliver the genes to the cornea. The hydrogels showed adequate light transparency, oxygen permeability, and swelling for use as contact lenses. Loading of viral vectors (rAAV-lacZ, rAAV-RFP, or rAAV-hIGF-I) was carried out at 4 °C to maintain viral vector titer. Release in culture medium was monitored by fluorescence with Cy3-rAAV-lacZ and AAV Titration ELISA. Transduction efficacy was tested through reporter genes lacZ and RFP in human bone marrow derived mesenchymal stem cells (hMSCs). lacZ was detected with X-Gal staining and quantified with Beta-Glo®, and RFP was monitored by fluorescence. The ability of rAAV-hIGF-I-loaded hydrogels to trigger cell proliferation in hMSCs was evaluated by immunohistochemistry. Finally, the ability of rAAV-lacZ-loaded hydrogels to transduce bovine cornea was confirmed through detection with X-Gal staining of β-galactosidase expressed within the tissue.
Collapse
Affiliation(s)
- Fernando Alvarez-Rivera
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+DFarma, Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (F.A.-R.); (L.D.-G.); (A.C.)
| | - Ana Rey-Rico
- Cell Therapy and Regenerative Medicine Unit, Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña, Campus de A Coruña, 15071 A Coruña, Spain;
| | - Jagadeesh K Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, 66421 Homburg, Germany; (J.K.V.); (M.C.)
| | - Luis Diaz-Gomez
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+DFarma, Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (F.A.-R.); (L.D.-G.); (A.C.)
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, 66421 Homburg, Germany; (J.K.V.); (M.C.)
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+DFarma, Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (F.A.-R.); (L.D.-G.); (A.C.)
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+DFarma, Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (F.A.-R.); (L.D.-G.); (A.C.)
- Correspondence: ; Tel.: +34-881815239
| |
Collapse
|
13
|
Sun K, Luo J, Guo J, Yao X, Jing X, Guo F. The PI3K/AKT/mTOR signaling pathway in osteoarthritis: a narrative review. Osteoarthritis Cartilage 2020; 28:400-409. [PMID: 32081707 DOI: 10.1016/j.joca.2020.02.027] [Citation(s) in RCA: 379] [Impact Index Per Article: 75.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/05/2020] [Accepted: 02/06/2020] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) is a complicated degenerative disease that affects whole joint tissue. Currently, apart from surgical approaches to treat late stage OA, effective treatments to reverse OA are not available. Thus, the mechanisms leading to OA, and more effective approaches to treat OA should be investigated. According to available evidence, the PI3K/AKT/mTOR signaling pathway is essential for normal metabolism of joint tissues, but is also involved in development of OA. To provide a wide viewpoint to roles of PI3K/AKT/mTOR signaling pathway in osteoarthritis, a comprehensive literature search was performed using PubMed terms 'PI3K OR AKT OR mTOR' and 'osteoarthritis'. This review highlights the role of PI3K/AKT/mTOR signaling in cartilage degradation, subchondral bone dysfunction, and synovial inflammation, and discusses how this signaling pathway affects development of the disease. We also summarize recent evidences of therapeutic approaches to treat OA by targeting the PI3K/AKT/mTOR pathway, and discuss potential challenges in developing these strategies for clinical treatment of OA.
Collapse
Affiliation(s)
- K Sun
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - J Luo
- The Center for Biomedical Research, The Tongji Hospital Research Building, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China.
| | - J Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - X Yao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - X Jing
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - F Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
14
|
Effects of rAAV-Mediated sox9 Overexpression on the Biological Activities of Human Osteoarthritic Articular Chondrocytes in Their Intrinsic Three-Dimensional Environment. J Clin Med 2019; 8:jcm8101637. [PMID: 31591319 PMCID: PMC6832991 DOI: 10.3390/jcm8101637] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/01/2019] [Accepted: 10/02/2019] [Indexed: 12/11/2022] Open
Abstract
Gene therapy for osteoarthritis offers powerful, long-lasting tools that are well adapted to treat such a slow, progressive disorder, especially those therapies based on the clinically adapted recombinant adeno-associated viral (rAAV) vectors. Here, we examined the ability of an rAAV construct carrying a therapeutic sequence for the cartilage-specific SOX9 transcription factor to modulate the phenotype of human osteoarthritic articular chondrocytes compared with normal chondrocytes in a three-dimensional environment where the cells are embedded in their extracellular matrix. Successful sox9 overexpression via rAAV was noted for at least 21 days, leading to the significant production of major matrix components (proteoglycans, type-II collagen) without affecting the proliferation of the cells, while the cells contained premature hypertrophic processes relative to control conditions (reporter rAAV-lacZ application, absence of vector treatment). These findings show the value of using rAAV to adjust the osteoarthritic phenotype when the chondrocytes are confined in their inherently altered environment and the possibility of impacting key cellular processes via gene therapy to remodel human osteoarthritic cartilage lesions.
Collapse
|
15
|
Remodeling of Human Osteochondral Defects via rAAV-Mediated Co-Overexpression of TGF-β and IGF-I from Implanted Human Bone Marrow-Derived Mesenchymal Stromal Cells. J Clin Med 2019; 8:jcm8091326. [PMID: 31466339 PMCID: PMC6781264 DOI: 10.3390/jcm8091326] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 08/22/2019] [Accepted: 08/26/2019] [Indexed: 12/01/2022] Open
Abstract
The application of chondrogenic gene sequences to human bone marrow-derived mesenchymal stromal cells (hMSCs) is an attractive strategy to activate the reparative activities of these cells as a means to enhance the processes of cartilage repair using indirect cell transplantation procedures that may improve the repopulation of cartilage lesions. In the present study, we examined the feasibility of co-delivering the highly competent transforming growth factor beta (TGF-β) with the insulin-like growth factor I (IGF-I) in hMSCs via recombinant adeno-associated virus (rAAV) vector-mediated gene transfer prior to implantation in a human model of osteochondral defect (OCD) ex vivo that provides a microenvironment similar to that of focal cartilage lesions. The successful co-overexpression of rAAV TGF-β/IGF-I in implanted hMSCs promoted the durable remodeling of tissue injury in human OCDs over a prolonged period of time (21 days) relative to individual gene transfer and the control (reporter lacZ gene) treatment, with enhanced levels of cell proliferation and matrix deposition (proteoglycans, type-II collagen) both in the lesions and at a distance, while hypertrophic, osteogenic, and catabolic processes could be advantageously delayed. These findings demonstrate the value of indirect, progenitor cell-based combined rAAV gene therapy to treat human focal cartilage defects in a natural environment as a basis for future clinical applications.
Collapse
|
16
|
Gonzalez-Fernandez T, Rathan S, Hobbs C, Pitacco P, Freeman FE, Cunniffe GM, Dunne NJ, McCarthy HO, Nicolosi V, O'Brien FJ, Kelly DJ. Pore-forming bioinks to enable spatio-temporally defined gene delivery in bioprinted tissues. J Control Release 2019; 301:13-27. [PMID: 30853527 DOI: 10.1016/j.jconrel.2019.03.006] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/02/2019] [Accepted: 03/06/2019] [Indexed: 12/17/2022]
Abstract
The regeneration of complex tissues and organs remains a major clinical challenge. With a view towards bioprinting such tissues, we developed a new class of pore-forming bioink to spatially and temporally control the presentation of therapeutic genes within bioprinted tissues. By blending sacrificial and stable hydrogels, we were able to produce bioinks whose porosity increased with time following printing. When combined with amphipathic peptide-based plasmid DNA delivery, these bioinks supported enhanced non-viral gene transfer to stem cells in vitro. By modulating the porosity of these bioinks, it was possible to direct either rapid and transient (pore-forming bioinks), or slower and more sustained (solid bioinks) transfection of host or transplanted cells in vivo. To demonstrate the utility of these bioinks for the bioprinting of spatially complex tissues, they were next used to zonally position stem cells and plasmids encoding for either osteogenic (BMP2) or chondrogenic (combination of TGF-β3, BMP2 and SOX9) genes within networks of 3D printed thermoplastic fibers to produce mechanically reinforced, gene activated constructs. In vivo, these bioprinted tissues supported the development of a vascularised, bony tissue overlaid by a layer of stable cartilage. When combined with multiple-tool biofabrication strategies, these gene activated bioinks can enable the bioprinting of a wide range of spatially complex tissues.
Collapse
Affiliation(s)
- T Gonzalez-Fernandez
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons, Ireland
| | - S Rathan
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - C Hobbs
- Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons, Ireland; School of Physics, Trinity College Dublin, Ireland; Centre for Research of Adaptive Nanostructures and Nanodevices, Trinity College Dublin, Ireland
| | - P Pitacco
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - F E Freeman
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - G M Cunniffe
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - N J Dunne
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons, Ireland; Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Ireland; School of Mechanical and Manufacturing Engineering, Dublin City University, Ireland; School of Pharmacy, Queen's University Belfast, UK
| | - H O McCarthy
- School of Pharmacy, Queen's University Belfast, UK
| | - V Nicolosi
- Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons, Ireland; School of Physics, Trinity College Dublin, Ireland; Centre for Research of Adaptive Nanostructures and Nanodevices, Trinity College Dublin, Ireland
| | - F J O'Brien
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons, Ireland; Tissue Engineering Research Group, Dept. of Anatomy, Royal College of Surgeons in, Ireland
| | - D J Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons, Ireland; Tissue Engineering Research Group, Dept. of Anatomy, Royal College of Surgeons in, Ireland.
| |
Collapse
|
17
|
Improved Chondrogenic Differentiation of rAAV SOX9-Modified Human MSCs Seeded in Fibrin-Polyurethane Scaffolds in a Hydrodynamic Environment. Int J Mol Sci 2018; 19:ijms19092635. [PMID: 30189664 PMCID: PMC6163252 DOI: 10.3390/ijms19092635] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 08/30/2018] [Accepted: 09/03/2018] [Indexed: 12/15/2022] Open
Abstract
The repair of focal articular cartilage defects remains a problem. Combining gene therapy with tissue engineering approaches using bone marrow-derived mesenchymal stem cells (MSCs) may allow the development of improved options for cartilage repair. Here, we examined whether a three-dimensional fibrin-polyurethane scaffold provides a favorable environment for the effective chondrogenic differentiation of human MSCs (hMSCs) overexpressing the cartilage-specific SOX9 transcription factor via recombinant adeno-associated virus (rAAV) -mediated gene transfer cultured in a hydrodynamic environment in vitro. Sustained SOX9 expression was noted in the constructs for at least 21 days, the longest time point evaluated. Such spatially defined SOX9 overexpression enhanced proliferative, metabolic, and chondrogenic activities compared with control (reporter lacZ gene transfer) treatment. Of further note, administration of the SOX9 vector was also capable of delaying premature hypertrophic and osteogenic differentiation in the constructs. This enhancement of chondrogenesis by spatially defined overexpression of human SOX9 demonstrate the potential benefits of using rAAV-modified hMSCs seeded in fibrin-polyurethane scaffolds as a promising approach for implantation in focal cartilage lesions to improve cartilage repair.
Collapse
|
18
|
Onset and Progression of Human Osteoarthritis-Can Growth Factors, Inflammatory Cytokines, or Differential miRNA Expression Concomitantly Induce Proliferation, ECM Degradation, and Inflammation in Articular Cartilage? Int J Mol Sci 2018; 19:ijms19082282. [PMID: 30081513 PMCID: PMC6121276 DOI: 10.3390/ijms19082282] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 07/22/2018] [Accepted: 08/01/2018] [Indexed: 12/30/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative whole joint disease, for which no preventative or therapeutic biological interventions are available. This is likely due to the fact that OA pathogenesis includes several signaling pathways, whose interactions remain unclear, especially at disease onset. Early OA is characterized by three key events: a rarely considered early phase of proliferation of cartilage-resident cells, in contrast to well-established increased synthesis, and degradation of extracellular matrix components and inflammation, associated with OA progression. We focused on the question, which of these key events are regulated by growth factors, inflammatory cytokines, and/or miRNA abundance. Collectively, we elucidated a specific sequence of the OA key events that are described best as a very early phase of proliferation of human articular cartilage (AC) cells and concomitant anabolic/catabolic effects that are accompanied by incipient pro-inflammatory effects. Many of the reviewed factors appeared able to induce one or two key events. Only one factor, fibroblast growth factor 2 (FGF2), is capable of concomitantly inducing all key events. Moreover, AC cell proliferation cannot be induced and, in fact, is suppressed by inflammatory signaling, suggesting that inflammatory signaling cannot be the sole inductor of all early OA key events, especially at disease onset.
Collapse
|
19
|
Rey-Rico A, Venkatesan JK, Schmitt G, Speicher-Mentges S, Madry H, Cucchiarini M. Effective Remodelling of Human Osteoarthritic Cartilage by sox9 Gene Transfer and Overexpression upon Delivery of rAAV Vectors in Polymeric Micelles. Mol Pharm 2018; 15:2816-2826. [DOI: 10.1021/acs.molpharmaceut.8b00331] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Ana Rey-Rico
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg D-66421, Germany
- Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña, Campus de A Coruña, 15071 A Coruña, Spain
| | - Jagadesh K. Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg D-66421, Germany
| | - Gertrud Schmitt
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg D-66421, Germany
| | - Susanne Speicher-Mentges
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg D-66421, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg D-66421, Germany
- Department of Orthopaedics and Orthopaedic Surgery, Saarland University Medical Center, Homburg D-66421, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg D-66421, Germany
| |
Collapse
|
20
|
Rey-Rico A, Venkatesan JK, Schmitt G, Concheiro A, Madry H, Alvarez-Lorenzo C, Cucchiarini M. rAAV-mediated overexpression of TGF-β via vector delivery in polymeric micelles stimulates the biological and reparative activities of human articular chondrocytes in vitro and in a human osteochondral defect model. Int J Nanomedicine 2017; 12:6985-6996. [PMID: 29033566 PMCID: PMC5614797 DOI: 10.2147/ijn.s144579] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Recombinant adeno-associated virus (rAAV) vectors are clinically adapted vectors to durably treat human osteoarthritis (OA). Controlled delivery of rAAV vectors via polymeric micelles was reported to enhance the temporal and spatial presentation of the vectors into their targets. Here, we tested the feasibility of delivering rAAV vectors via poly (ethylene oxide) (PEO) and poly (propylene oxide) (PPO) (poloxamer and poloxamine) polymeric micelles as a means to overexpress the therapeutic factor transforming growth factor-beta (TGF-β) in human OA chondrocytes and in experimental human osteochondral defects. Application of rAAV-human transforming growth factor-beta using such micelles increased the levels of TGF-β transgene expression compared with free vector treatment. Overexpression of TGF-β with these systems resulted in higher proteoglycan deposition and increased cell numbers in OA chondrocytes. In osteochondral defect cultures, a higher deposition of type-II collagen and reduced hypertrophic events were noted. Delivery of therapeutic rAAV vectors via PEO-PPO-PEO micelles may provide potential tools to remodel human OA cartilage.
Collapse
Affiliation(s)
- Ana Rey-Rico
- Center of Experimental Orthopedics, Saarland University Medical Center, Homburg, Germany
| | - Jagadeesh K Venkatesan
- Center of Experimental Orthopedics, Saarland University Medical Center, Homburg, Germany
| | - Gertrud Schmitt
- Center of Experimental Orthopedics, Saarland University Medical Center, Homburg, Germany
| | - Angel Concheiro
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, R+ DPharma Group (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Henning Madry
- Center of Experimental Orthopedics, Saarland University Medical Center, Homburg, Germany.,Department of Orthopedics and Orthopedic Surgery, Saarland University Medical Center, Homburg, Germany
| | - Carmen Alvarez-Lorenzo
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, R+ DPharma Group (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Magali Cucchiarini
- Center of Experimental Orthopedics, Saarland University Medical Center, Homburg, Germany
| |
Collapse
|
21
|
Nieminen HJ, Barreto G, Finnilä MA, García-Pérez A, Salmi A, Ranjan S, Eklund KK, Pritzker KPH, Saarakkala S, Hæggström E. Laser-ultrasonic delivery of agents into articular cartilage. Sci Rep 2017; 7:3991. [PMID: 28638116 PMCID: PMC5479804 DOI: 10.1038/s41598-017-04293-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 05/12/2017] [Indexed: 11/09/2022] Open
Abstract
Research is ongoing to develop drug therapies to manage osteoarthritis (OA) and articular cartilage (AC) injuries. However, means to deliver drug to localized AC lesions are highly limited and not clinically available. This study investigates the capability of laser ultrasound (laser-induced plasma sound source) to deliver agents (methylene blue, MB, in PBS) into bovine AC. Treatment samples (n = 10) were immersed in MB solution simultaneously with LU exposure, while adjacent control 1 tissue (n = 10) was pre-treated with LU followed by immersion in MB and adjacent control 2 tissue (n = 10) was only immersed in MB. AC exposed (n = 22) or not exposed (n = 27) to LU were characterized for anomalies in structure, composition, viability or RNA expression. Optically detected MB content was significantly (p < 0.01) higher in treatment samples up to a depth of 500 µm from AC surface as compared to controls. No major unwanted short-term effects on AC structure, proteoglycan or collagen contents, chondrocyte viability or RNA expression levels were detected. In conclusion, LU can deliver agents into AC without major short-term concerns on safety. LU could reveal new strategies for the development of localized drug therapies in AC.
Collapse
Affiliation(s)
- Heikki J Nieminen
- Electronics Research Laboratory, Department of Physics, University of Helsinki, Helsinki, Finland.
- Research Group of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
| | - Gonçalo Barreto
- Orton Orthopaedic Hospital and Research Institute, Invalid Foundation, Helsinki, Finland
- Department of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikko A Finnilä
- Research Group of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - Alejandro García-Pérez
- Electronics Research Laboratory, Department of Physics, University of Helsinki, Helsinki, Finland
- Department of Electronic Engineering, Higher Technological Institute of Poza Rica, Poza Rica, México, USA
| | - Ari Salmi
- Electronics Research Laboratory, Department of Physics, University of Helsinki, Helsinki, Finland
| | - Sanjeev Ranjan
- Laboratory of Radiochemistry, Department of Chemistry, University of Helsinki, Helsinki, Finland
| | - Kari K Eklund
- Department of Rheumatology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Kenneth P H Pritzker
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, Mount Sinai Hospital, Toronto, Canada
| | - Simo Saarakkala
- Research Group of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
| | - Edward Hæggström
- Electronics Research Laboratory, Department of Physics, University of Helsinki, Helsinki, Finland
| |
Collapse
|
22
|
Gonzalez-Fernandez T, Sathy B, Hobbs C, Cunniffe G, McCarthy H, Dunne N, Nicolosi V, O'Brien F, Kelly D. Mesenchymal stem cell fate following non-viral gene transfection strongly depends on the choice of delivery vector. Acta Biomater 2017; 55:226-238. [PMID: 28363788 DOI: 10.1016/j.actbio.2017.03.044] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/06/2017] [Accepted: 03/27/2017] [Indexed: 12/22/2022]
Abstract
Controlling the phenotype of mesenchymal stem cells (MSCs) through the delivery of regulatory genes is a promising strategy in tissue engineering (TE). Essential to effective gene delivery is the choice of gene carrier. Non-viral delivery vectors have been extensively used in TE, however their intrinsic effects on MSC differentiation remain poorly understood. The objective of this study was to investigate the influence of three different classes of non-viral gene delivery vectors: (1) cationic polymers (polyethylenimine, PEI), (2) inorganic nanoparticles (nanohydroxyapatite, nHA) and (3) amphipathic peptides (RALA peptide) on modulating stem cell fate after reporter and therapeutic gene delivery. Despite facilitating similar reporter gene transfection efficiencies, these nanoparticle-based vectors had dramatically different effects on MSC viability, cytoskeletal morphology and differentiation. After reporter gene delivery (pGFP or pLUC), the nHA and RALA vectors supported an elongated MSC morphology, actin stress fibre formation and the development of mature focal adhesions, while cells appeared rounded and less tense following PEI transfection. These changes in MSC morphology correlated with enhanced osteogenesis following nHA and RALA transfection and adipogenesis following PEI transfection. When therapeutic genes encoding for transforming growth factor beta 3 (TGF-β3) and/or bone morphogenic protein 2 (BMP2) were delivered to MSCs, nHA promoted osteogenesis in 2D culture and the development of an endochondral phenotype in 3D culture, while RALA was less osteogenic and appeared to promote a more stable hyaline cartilage-like phenotype. In contrast, PEI failed to induce robust osteogenesis or chondrogenesis of MSCs, despite effective therapeutic protein production. Taken together, these results demonstrate that the differentiation of MSCs through the application of non-viral gene delivery strategies depends not only on the gene delivered, but also on the gene carrier itself. STATEMENT OF SIGNIFICANCE Nanoparticle-based non-viral gene delivery vectors have been extensively used in regenerative medicine, however their intrinsic effects on mesenchymal stem cell (MSC) differentiation remain poorly understood. This paper demonstrates that different classes of commonly used non-viral vectors are not inert and they have a strong effect on cell morphology, stress fiber formation and gene transcription in MSCs, which in turn modulates their capacity to differentiate towards osteogenic, adipogenic and chondrogenic lineages. These results also point to the need for careful and tissue-specific selection of nanoparticle-based delivery vectors to prevent undesired phenotypic changes and off-target effects when delivering therapeutic genes to damaged or diseased tissues.
Collapse
|
23
|
Rey-Rico A, Babicz H, Madry H, Concheiro A, Alvarez-Lorenzo C, Cucchiarini M. Supramolecular polypseudorotaxane gels for controlled delivery of rAAV vectors in human mesenchymal stem cells for regenerative medicine. Int J Pharm 2017; 531:492-503. [PMID: 28552768 DOI: 10.1016/j.ijpharm.2017.05.050] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 05/17/2017] [Accepted: 05/22/2017] [Indexed: 12/12/2022]
Abstract
The aim of this work was to investigate, for the first time, the possibility of using supramolecular polypseudorotaxane gels as scaffolds that can durably deliver rAAV vectors for applications in cartilage regeneration. Dispersions of Pluronic® F68 (PF68) or Tetronic® 908 (T908) containing either hyaluronic acid (HA) or chondroitin sulfate (CS) were prepared in PBS. Then, alpha-cyclodextrin (αCD) was added to some dispersions to form polypseudorotaxane gels. Polysaccharides and αCD reinforced the viscoelasticity of the gels, which could withstand autoclaving without changes. In vitro release of rAAV vectors and subsequent transduction of human mesenchymal stem cells (hMSCs) by rAAV vectors from the release medium and from gels in direct contact with the cells were investigated. Compared with free vectors, the gels provided higher levels of transgene expression. CS (or HA)/PF68/αCD gels rapidly released rAAV vectors while CS (or HA)/T908/αCD gels provided sustained release probably due to different interactions with the viral vectors. Incorporation of αCD into CS (or HA)/PF68 gels resulted on higher rAAV concentrations and sustained levels of transgene expression over time. HA increased the bioactivity and cytocompatibility of the gels, especially those based on T908. Overall, combining rAAV gene transfer with polypseudorotaxane gels may provide new, promising tools for human tissue engineering and regenerative medicine strategies.
Collapse
Affiliation(s)
- Ana Rey-Rico
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg, Germany.
| | - Heiko Babicz
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg, Germany; Department of Orthopaedics and Orthopaedic Surgery, Saarland University Medical Center, Homburg, Germany
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, R+DPharma Group (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15872 Santiago de Compostela, Spain
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, R+DPharma Group (GI-1645), Facultad de Farmacia and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15872 Santiago de Compostela, Spain.
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg, Germany
| |
Collapse
|
24
|
Rey-Rico A, Cucchiarini M. Recent tissue engineering-based advances for effective rAAV-mediated gene transfer in the musculoskeletal system. Bioengineered 2017; 7:175-88. [PMID: 27221233 DOI: 10.1080/21655979.2016.1187347] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Musculoskeletal tissues are diverse and significantly different in their ability to repair upon injury. Current treatments often fail to reproduce the natural functions of the native tissue, leading to an imperfect healing. Gene therapy might improve the repair of tissues by providing a temporarily and spatially defined expression of the therapeutic gene(s) at the site of the injury. Several gene transfer vehicles have been developed to modify various human cells and tissues from musculoskeletal system among which the non-pathogenic, effective, and relatively safe recombinant adeno-associated viral (rAAV) vectors that have emerged as the preferred gene delivery system to treat human disorders. Adapting tissue engineering platforms to gene transfer approaches mediated by rAAV vectors is an attractive tool to circumvent both the limitations of the current therapeutic options to promote an effective healing of the tissue and the natural obstacles from these clinically adapted vectors to achieve an efficient and durable gene expression of the therapeutic sequences within the lesions.
Collapse
Affiliation(s)
- Ana Rey-Rico
- a Center of Experimental Orthopaedics , Saarland University Medical Center , Homburg/Saar , Germany
| | - Magali Cucchiarini
- a Center of Experimental Orthopaedics , Saarland University Medical Center , Homburg/Saar , Germany
| |
Collapse
|
25
|
Insights on Molecular Mechanisms of Chondrocytes Death in Osteoarthritis. Int J Mol Sci 2016; 17:ijms17122146. [PMID: 27999417 PMCID: PMC5187946 DOI: 10.3390/ijms17122146] [Citation(s) in RCA: 250] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 12/05/2016] [Accepted: 12/12/2016] [Indexed: 12/21/2022] Open
Abstract
Osteoarthritis (OA) is a joint pathology characterized by progressive cartilage degradation. Medical care is mainly based on alleviating pain symptoms. Compelling studies report the presence of empty lacunae and hypocellularity in cartilage with aging and OA progression, suggesting that chondrocyte cell death occurs and participates to OA development. However, the relative contribution of apoptosis per se in OA pathogenesis appears complex to evaluate. Indeed, depending on technical approaches, OA stages, cartilage layers, animal models, as well as in vivo or in vitro experiments, the percentage of apoptosis and cell death types can vary. Apoptosis, chondroptosis, necrosis, and autophagic cell death are described in this review. The question of cell death causality in OA progression is also addressed, as well as the molecular pathways leading to cell death in response to the following inducers: Fas, Interleukin-1β (IL-1β), Tumor Necrosis factor-α (TNF-α), leptin, nitric oxide (NO) donors, and mechanical stresses. Furthermore, the protective role of autophagy in chondrocytes is highlighted, as well as its decline during OA progression, enhancing chondrocyte cell death; the transition being mainly controlled by HIF-1α/HIF-2α imbalance. Finally, we have considered whether interfering in chondrocyte apoptosis or promoting autophagy could constitute therapeutic strategies to impede OA progression.
Collapse
|
26
|
Coricor G, Serra R. TGF-β regulates phosphorylation and stabilization of Sox9 protein in chondrocytes through p38 and Smad dependent mechanisms. Sci Rep 2016; 6:38616. [PMID: 27929080 PMCID: PMC5144132 DOI: 10.1038/srep38616] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 11/11/2016] [Indexed: 12/25/2022] Open
Abstract
Members of the TGF-β superfamily are important regulators of chondrocyte function. Sox9, a key transcriptional regulator of chondrogenesis, is required for TGF-β-mediated regulation of specific cartilage genes. TGF-β can signal through a canonical, Smad-mediated pathway or non-conical pathways, including p38. Here we show that both pathways are activated in chondrocytes after treatment with TGF-β and that TGF-β stabilizes Sox9 protein and increases phosphorylation of Sox9. Mutagenesis of potential serine phosphorylation sites on Sox9 was used to demonstrate that serine 211 is required to maintain normal basal levels of Sox9 as well as mediate increased Sox9 levels in response to TGF-β. The serine 211 site is in a motif that is targeted by p38 kinase. We used siRNA and pharmacological agents to show that p38 and Smad3 independently regulate the phosphorylation and stability of Sox9. Previously, we demonstrated that Papss2 is a downstream transcriptional target of Sox9 and TGF-β. Here we show that p38 is required for TGF-β-mediated regulation of Papss2 mRNA. Together the results suggest a new mechanism for TGF-β-mediated gene regulation in chondrocytes via p38 and phosphorylation and stabilization of Sox9. Understanding how TGF-β regulates Sox9 may lead to identification of therapeutic targets for OA.
Collapse
Affiliation(s)
- George Coricor
- University of Alabama at Birmingham, Department of Cell, Developmental, and Integrative Biology, Birmingham, Alabama, 35294-0005, USA
| | - Rosa Serra
- University of Alabama at Birmingham, Department of Cell, Developmental, and Integrative Biology, Birmingham, Alabama, 35294-0005, USA
| |
Collapse
|
27
|
Tao K, Rey-Rico A, Frisch J, Venkatesan JK, Schmitt G, Madry H, Lin J, Cucchiarini M. rAAV-mediated combined gene transfer and overexpression of TGF-β and SOX9 remodels human osteoarthritic articular cartilage. J Orthop Res 2016; 34:2181-2190. [PMID: 26970525 DOI: 10.1002/jor.23228] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 03/06/2016] [Indexed: 02/04/2023]
Abstract
Direct administration of therapeutic candidate gene sequences using the safe and effective recombinant adeno-associated virus (rAAV) vectors is a promising strategy to stimulate the biologic activities of articular chondrocytes as an adapted tool to treat human osteoarthritic (OA) cartilage. In the present study, we developed a combined gene transfer approach based on the co-delivery of the pleiotropic transformation growth factor beta (TGF-β) with the specific transcription factor SOX9 via rAAV to human normal and OA chondrocytes in vitro and cartilage explants in situ in light of the mitogenic and pro-anabolic properties of these factors. Effective, durable co-overexpression of TGF-β and SOX9 significantly enhanced the levels of cell proliferation both in human normal and OA chondrocytes and cartilage explants over an extended period of time (21 days), while stimulating the biosynthesis of key matrix components (proteoglycans, type-II collagen) compared with control conditions (reporter lacZ gene transfer, absence of vector treatment). Of further note, expression of hypertrophic type-X collagen significantly decreased following co-treatment by the candidate vectors. The present findings show the value of combining the transfer and expression of potent candidate factors in human OA cartilage as a means to re-establish essential features of normal cartilage and counteract the pathological shift of homeostasis. These observations support the concept of developing dual therapeutic rAAV gene transfer strategies as future, adapted tools for the direct treatment of human OA. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:2181-2190, 2016.
Collapse
Affiliation(s)
- Ke Tao
- Institute of Arthritis, Peking University People's Hospital, Beijing, 100044, People's Republic of China
- Peking University Health Science Center, Beijing, 100191, People's Republic of China
- Center of Experimental Orthopedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Ana Rey-Rico
- Center of Experimental Orthopedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Janina Frisch
- Center of Experimental Orthopedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Jagadeesh K Venkatesan
- Center of Experimental Orthopedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Gertrud Schmitt
- Center of Experimental Orthopedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopedics, Saarland University Medical Center, Homburg/Saar, Germany
- Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg/Saar, Germany
| | - Jianhao Lin
- Institute of Arthritis, Peking University People's Hospital, Beijing, 100044, People's Republic of China
- Peking University Health Science Center, Beijing, 100191, People's Republic of China
| | - Magali Cucchiarini
- Center of Experimental Orthopedics, Saarland University Medical Center, Homburg/Saar, Germany
| |
Collapse
|
28
|
Frisch J, Cucchiarini M. Gene- and Stem Cell-Based Approaches to Regulate Hypertrophic Differentiation in Articular Cartilage Disorders. Stem Cells Dev 2016; 25:1495-1512. [DOI: 10.1089/scd.2016.0106] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Janina Frisch
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Homburg, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Homburg, Germany
| |
Collapse
|
29
|
Ondrésik M, Azevedo Maia FR, da Silva Morais A, Gertrudes AC, Dias Bacelar AH, Correia C, Gonçalves C, Radhouani H, Amandi Sousa R, Oliveira JM, Reis RL. Management of knee osteoarthritis. Current status and future trends. Biotechnol Bioeng 2016; 114:717-739. [DOI: 10.1002/bit.26182] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 07/13/2016] [Accepted: 09/09/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Marta Ondrésik
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics; Universidade do Minho, Headquarters of the European Institute Regenerative Medicine; AvePark 4806-909, Caldas das Taipas Guimaraes Portugal
- ICVS/3B's-PT Government Associated Laboratory; Braga/Guimaraes Portugal
| | - Fatima R. Azevedo Maia
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics; Universidade do Minho, Headquarters of the European Institute Regenerative Medicine; AvePark 4806-909, Caldas das Taipas Guimaraes Portugal
- ICVS/3B's-PT Government Associated Laboratory; Braga/Guimaraes Portugal
| | - Alain da Silva Morais
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics; Universidade do Minho, Headquarters of the European Institute Regenerative Medicine; AvePark 4806-909, Caldas das Taipas Guimaraes Portugal
- ICVS/3B's-PT Government Associated Laboratory; Braga/Guimaraes Portugal
- Stemmatters, Biotecnologia e Medicina Regenerativa SA; Guimaraes Portugal
| | - Ana C. Gertrudes
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics; Universidade do Minho, Headquarters of the European Institute Regenerative Medicine; AvePark 4806-909, Caldas das Taipas Guimaraes Portugal
- ICVS/3B's-PT Government Associated Laboratory; Braga/Guimaraes Portugal
- Stemmatters, Biotecnologia e Medicina Regenerativa SA; Guimaraes Portugal
| | - Ana H. Dias Bacelar
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics; Universidade do Minho, Headquarters of the European Institute Regenerative Medicine; AvePark 4806-909, Caldas das Taipas Guimaraes Portugal
- ICVS/3B's-PT Government Associated Laboratory; Braga/Guimaraes Portugal
- Stemmatters, Biotecnologia e Medicina Regenerativa SA; Guimaraes Portugal
| | - Cristina Correia
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics; Universidade do Minho, Headquarters of the European Institute Regenerative Medicine; AvePark 4806-909, Caldas das Taipas Guimaraes Portugal
- ICVS/3B's-PT Government Associated Laboratory; Braga/Guimaraes Portugal
- Stemmatters, Biotecnologia e Medicina Regenerativa SA; Guimaraes Portugal
| | - Cristiana Gonçalves
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics; Universidade do Minho, Headquarters of the European Institute Regenerative Medicine; AvePark 4806-909, Caldas das Taipas Guimaraes Portugal
- ICVS/3B's-PT Government Associated Laboratory; Braga/Guimaraes Portugal
- Stemmatters, Biotecnologia e Medicina Regenerativa SA; Guimaraes Portugal
| | - Hajer Radhouani
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics; Universidade do Minho, Headquarters of the European Institute Regenerative Medicine; AvePark 4806-909, Caldas das Taipas Guimaraes Portugal
- ICVS/3B's-PT Government Associated Laboratory; Braga/Guimaraes Portugal
- Stemmatters, Biotecnologia e Medicina Regenerativa SA; Guimaraes Portugal
| | - Rui Amandi Sousa
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics; Universidade do Minho, Headquarters of the European Institute Regenerative Medicine; AvePark 4806-909, Caldas das Taipas Guimaraes Portugal
- ICVS/3B's-PT Government Associated Laboratory; Braga/Guimaraes Portugal
- Stemmatters, Biotecnologia e Medicina Regenerativa SA; Guimaraes Portugal
| | - Joaquim M. Oliveira
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics; Universidade do Minho, Headquarters of the European Institute Regenerative Medicine; AvePark 4806-909, Caldas das Taipas Guimaraes Portugal
- ICVS/3B's-PT Government Associated Laboratory; Braga/Guimaraes Portugal
| | - Rui L. Reis
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics; Universidade do Minho, Headquarters of the European Institute Regenerative Medicine; AvePark 4806-909, Caldas das Taipas Guimaraes Portugal
- ICVS/3B's-PT Government Associated Laboratory; Braga/Guimaraes Portugal
| |
Collapse
|
30
|
Frisch J, Orth P, Venkatesan JK, Rey‐Rico A, Schmitt G, Kohn D, Madry H, Cucchiarini M. Genetic Modification of Human Peripheral Blood Aspirates Using Recombinant Adeno-Associated Viral Vectors for Articular Cartilage Repair with a Focus on Chondrogenic Transforming Growth Factor-β Gene Delivery. Stem Cells Transl Med 2016; 6:249-260. [PMID: 28170175 PMCID: PMC5442727 DOI: 10.5966/sctm.2016-0149] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/28/2016] [Indexed: 01/13/2023] Open
Abstract
Transplantation of genetically modified peripheral blood aspirates that carry chondrogenically competent progenitor cells may offer new, convenient tools to treat articular cartilage lesions compared with the more complex and invasive application of bone marrow concentrates or of bone marrow‐derived mesenchymal stem cells. Here, we show that recombinant adeno‐associated viral (rAAV) vectors are powerful gene vehicles capable of successfully targeting primary human peripheral blood aspirates in a stable and safe manner, allowing for an efficient and long‐term transgene expression in such samples (up to 63 days with use of a lacZ reporter gene and for at least 21 days with application of the pleiotropic, chondrogenic factor transforming growth factor‐β [TGF‐β]). rAAV‐mediated overexpression of TGF‐β enhanced both the proliferative and metabolic properties of the peripheral blood aspirates, also increasing the chondrogenic differentiation processes in these samples. Hypertrophy and osteogenic differentiation events were also activated by production of TGF‐β via rAAV, suggesting that translation of the current approach in vivo will probably require close regulation of expression of this candidate gene. However, these results support the concept of directly modifying peripheral blood as a novel approach to conveniently treat articular cartilage lesions in patients. Stem Cells Translational Medicine2017;6:249–260
Collapse
Affiliation(s)
- Janina Frisch
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Patrick Orth
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
- Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg/Saar, Germany
| | | | - Ana Rey‐Rico
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Gertrud Schmitt
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Dieter Kohn
- Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg/Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
- Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg/Saar, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| |
Collapse
|
31
|
Rey-Rico A, Frisch J, Venkatesan JK, Schmitt G, Rial-Hermida I, Taboada P, Concheiro A, Madry H, Alvarez-Lorenzo C, Cucchiarini M. PEO-PPO-PEO Carriers for rAAV-Mediated Transduction of Human Articular Chondrocytes in Vitro and in a Human Osteochondral Defect Model. ACS APPLIED MATERIALS & INTERFACES 2016; 8:20600-20613. [PMID: 27404480 DOI: 10.1021/acsami.6b06509] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Gene therapy is an attractive strategy for the durable treatment of human osteoarthritis (OA), a gradual, irreversible joint disease. Gene carriers based on the small human adeno-associated virus (AAV) exhibit major efficacy in modifying damaged human articular cartilage in situ over extended periods of time. Yet, clinical application of recombinant AAV (rAAV) vectors remains complicated by the presence of neutralizing antibodies against viral capsid elements in a majority of patients. The goal of this study was to evaluate the feasibility of delivering rAAV vectors to human OA chondrocytes in vitro and in an experimental model of osteochondral defect via polymeric micelles to protect gene transfer from experimental neutralization. Interaction of rAAV with micelles of linear (poloxamer PF68) or X-shaped (poloxamine T908) poly(ethylene oxide) (PEO) and poly(propylene oxide) (PPO) copolymers (PEO-PPO-PEO micelles) was characterized by means of isothermal titration calorimetry. Micelle encapsulation allowed an increase in both the stability and bioactivity of rAAV vectors and promoted higher levels of safe transgene (lacZ) expression both in vitro and in experimental osteochondral defects compared with that of free vector treatment without detrimental effects on the biological activity of the cells or their phenotype. Remarkably, protection against antibody neutralization was also afforded when delivering rAAV via PEO-PPO-PEO micelles in all systems evaluated, especially when using T908. Altogether, these findings show the potential of PEO-PPO-PEO micelles as effective tools to improve current gene-based treatments for human OA.
Collapse
Affiliation(s)
- Ana Rey-Rico
- Center of Experimental Orthopaedics, Saarland University Medical Center , Homburg, Germany
| | - Janina Frisch
- Center of Experimental Orthopaedics, Saarland University Medical Center , Homburg, Germany
| | | | - Gertrud Schmitt
- Center of Experimental Orthopaedics, Saarland University Medical Center , Homburg, Germany
| | - Isabel Rial-Hermida
- Departamento de Farmacia y Tecnología Farmacéutica, R+DPharma Group (GI-1645), Facultad de Farmacia, Universidade de Santiago de Compostela , Santiago de Compostela, Spain
| | - Pablo Taboada
- Departamento de Física de la Materia Condensada, Facultad de Física, Universidade de Santiago de Compostela , Santiago de Compostela, Spain
| | - Angel Concheiro
- Departamento de Farmacia y Tecnología Farmacéutica, R+DPharma Group (GI-1645), Facultad de Farmacia, Universidade de Santiago de Compostela , Santiago de Compostela, Spain
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center , Homburg, Germany
- Department of Orthopaedics and Orthopaedic Surgery, Saarland University Medical Center , Homburg, Germany
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacia y Tecnología Farmacéutica, R+DPharma Group (GI-1645), Facultad de Farmacia, Universidade de Santiago de Compostela , Santiago de Compostela, Spain
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center , Homburg, Germany
| |
Collapse
|
32
|
Frisch J, Rey-Rico A, Venkatesan JK, Schmitt G, Madry H, Cucchiarini M. TGF-β gene transfer and overexpression via rAAV vectors stimulates chondrogenic events in human bone marrow aspirates. J Cell Mol Med 2016; 20:430-40. [PMID: 26808466 PMCID: PMC4759465 DOI: 10.1111/jcmm.12774] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 12/01/2015] [Indexed: 12/12/2022] Open
Abstract
Genetic modification of marrow concentrates may provide convenient approaches to enhance the chondrogenic differentiation processes and improve the repair capacities in sites of cartilage defects following administration in the lesions. Here, we provided clinically adapted recombinant adeno‐associated virus (rAAV) vectors to human bone marrow aspirates to promote the expression of the potent transforming growth factor beta (TGF‐β) as a means to regulate the biological and chondrogenic activities in the samples in vitro. Successful TGF‐β gene transfer and expression viarAAV was reached relative to control (lacZ) treatment (from 511.1 to 16.1 pg rhTGF‐β/mg total proteins after 21 days), allowing to durably enhance the levels of cell proliferation, matrix synthesis, and chondrogenic differentiation. Strikingly, in the conditions applied here, application of the candidate TGF‐β vector was also capable of reducing the hypertrophic and osteogenic differentiation processes in the aspirates, showing the potential benefits of using this particular vector to directly modify marrow concentrates to generate single‐step, effective approaches that aim at improving articular cartilage repair in vivo.
Collapse
Affiliation(s)
- Janina Frisch
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Ana Rey-Rico
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | | | - Gertrud Schmitt
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany.,Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg/Saar, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| |
Collapse
|
33
|
Díaz-Rodríguez P, Rey-Rico A, Madry H, Landin M, Cucchiarini M. Effective genetic modification and differentiation of hMSCs upon controlled release of rAAV vectors using alginate/poloxamer composite systems. Int J Pharm 2015; 496:614-26. [PMID: 26556623 DOI: 10.1016/j.ijpharm.2015.11.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 11/03/2015] [Accepted: 11/04/2015] [Indexed: 02/02/2023]
Abstract
Viral vectors are common tools in gene therapy to deliver foreign therapeutic sequences in a specific target population via their natural cellular entry mechanisms. Incorporating such vectors in implantable systems may provide strong alternatives to conventional gene transfer procedures. The goal of the present study was to generate different hydrogel structures based on alginate (AlgPH155) and poloxamer PF127 as new systems to encapsulate and release recombinant adeno-associated viral (rAAV) vectors. Inclusion of rAAV in such polymeric capsules revealed an influence of the hydrogel composition and crosslinking temperature upon the vector release profiles, with alginate (AlgPH155) structures showing the fastest release profiles early on while over time vector release was more effective from AlgPH155+PF127 [H] capsules crosslinked at a high temperature (50°C). Systems prepared at room temperature (AlgPH155+PF127 [C]) allowed instead to achieve a more controlled release profile. When tested for their ability to target human mesenchymal stem cells, the different systems led to high transduction efficiencies over time and to gene expression levels in the range of those achieved upon direct vector application, especially when using AlgPH155+PF127 [H]. No detrimental effects were reported on either cell viability or on the potential for chondrogenic differentiation. Inclusion of PF127 in the capsules was also capable of delaying undesirable hypertrophic cell differentiation. These findings are of promising value for the further development of viral vector controlled release strategies.
Collapse
Affiliation(s)
- P Díaz-Rodríguez
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany; Departamento de Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Universidad de Santiago de Compostela, Spain
| | - A Rey-Rico
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - H Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany; Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg/Saar, Germany
| | - M Landin
- Departamento de Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Universidad de Santiago de Compostela, Spain
| | - M Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany.
| |
Collapse
|
34
|
Nieminen HJ, Ylitalo T, Suuronen JP, Rahunen K, Salmi A, Saarakkala S, Serimaa R, Hæggström E. Delivering Agents Locally into Articular Cartilage by Intense MHz Ultrasound. ULTRASOUND IN MEDICINE & BIOLOGY 2015; 41:2259-2265. [PMID: 25922135 PMCID: PMC4587538 DOI: 10.1016/j.ultrasmedbio.2015.03.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 03/18/2015] [Accepted: 03/27/2015] [Indexed: 06/04/2023]
Abstract
There is no cure for osteoarthritis. Current drug delivery relies on systemic delivery or injections into the joint. Because articular cartilage (AC) degeneration can be local and drug exposure outside the lesion can cause adverse effects, localized drug delivery could permit new drug treatment strategies. We investigated whether intense megahertz ultrasound (frequency: 1.138 MHz, peak positive pressure: 2.7 MPa, Ispta: 5 W/cm(2), beam width: 5.7 mm at -6 dB, duty cycle: 5%, pulse repetition frequency: 285 Hz, mechanical index: 1.1) can deliver agents into AC without damaging it. Using ultrasound, we delivered a drug surrogate down to a depth corresponding to 53% depth of the AC thickness without causing histologically detectable damage to the AC. This may be important because early osteoarthritis typically exhibits histopathologic changes in the superficial AC. In conclusion, we identify intense megahertz ultrasound as a technique that potentially enables localized non-destructive delivery of osteoarthritis drugs or drug carriers into articular cartilage.
Collapse
Affiliation(s)
| | - Tuomo Ylitalo
- Department of Physics, University of Helsinki, Helsinki, Finland; Research Center for Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | | | - Krista Rahunen
- Research Center for Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Ari Salmi
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Simo Saarakkala
- Research Center for Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland; Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
| | - Ritva Serimaa
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Edward Hæggström
- Department of Physics, University of Helsinki, Helsinki, Finland
| |
Collapse
|
35
|
Rey-Rico A, Venkatesan JK, Frisch J, Schmitt G, Monge-Marcet A, Lopez-Chicon P, Mata A, Semino C, Madry H, Cucchiarini M. Effective and durable genetic modification of human mesenchymal stem cells via controlled release of rAAV vectors from self-assembling peptide hydrogels with a maintained differentiation potency. Acta Biomater 2015; 18:118-27. [PMID: 25712390 DOI: 10.1016/j.actbio.2015.02.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 02/09/2015] [Accepted: 02/13/2015] [Indexed: 10/24/2022]
Abstract
Controlling the release of recombinant adeno-associated virus (rAAV) vectors from biocompatible materials is a novel, attractive approach to increase the residence time and effectiveness of a gene carrier at a defined target site. Self-assembling peptides have an ability to form stable hydrogels and encapsulate cells upon exposure to physiological pH and ionic strength. Here, we examined the capacity of the peptide hydrogel RAD16-I in a pure (RAD) form or combined with hyaluronic acid (RAD-HA) to release rAAV vectors as a means to genetically modify primary human bone marrow-derived mesenchymal stem cells (hMSCs), a potent source of cells for regenerative medicine. Specifically, we demonstrate the ability of the systems to efficiently encapsulate and release rAAV vectors in a sustained, controlled manner for the effective transduction of hMSCs (up to 80%) without deleterious effects on cell viability (up to 100%) or on their potential for chondrogenic differentiation over time (up to 21days). The present study demonstrates that RAD16-I is an advantageous material with tunable properties to control the release of rAAV vectors as a promising tool to develop new, improved therapeutic approaches for tissue engineering in vivo.
Collapse
|
36
|
Cucchiarini M, Schmidt K, Frisch J, Kohn D, Madry H. Overexpression of TGF-β via rAAV-Mediated Gene Transfer Promotes the Healing of Human Meniscal Lesions Ex Vivo on Explanted Menisci. Am J Sports Med 2015; 43:1197-205. [PMID: 25646364 DOI: 10.1177/0363546514567063] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Direct application of therapeutic gene vectors derived from the adeno-associated virus (AAV) might be beneficial to improve the healing of meniscal tears. PURPOSE To test the ability of recombinant AAV (rAAV) to overexpress the potent transforming growth factor-β (TGF-β) in primary cultures of human meniscal fibrochondrocytes, in human meniscal explants, and in experimental human meniscal lesions as a new tool to enhance meniscal repair. STUDY DESIGN Controlled laboratory study. METHODS The effects of the candidate treatment on the proliferative and metabolic activities of meniscal cells were monitored in vitro for up to 21 days and in situ in intact and injured human menisci for up to 15 days using biochemical, immunohistochemical, histological, and histomorphometric analyses. RESULTS Efficient production of TGF-β via rAAV was achieved in vitro and in situ, both in the intact and injured meniscus. Application of the rAAV TGF-β vector stimulated the levels of cell proliferation and matrix synthesis (type I collagen) compared with control gene transfer in all systems tested, especially in situ in regions of poor healing capacity and in sites of meniscal injury. No adverse effects of the candidate treatment were observed at the level of osseous differentiation, as tested by immunodetection of type X collagen. Most remarkably, a significant reduction of the amplitude of meniscal tears was noted after TGF-β treatment, an effect that was associated with increased expression levels of the α-smooth muscle actin contractile marker. CONCLUSION Overexpression of TGF-β via rAAV gene transfer is capable of modulating the reparative activities of human meniscal cells, allowing for the healing of meniscal lesions by convenient injection in sites of injury. CLINICAL RELEVANCE Direct gene-based approaches using rAAV have strong potential to develop new therapeutic options that aim at treating human meniscal defects.
Collapse
Affiliation(s)
- Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Katharina Schmidt
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Janina Frisch
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Dieter Kohn
- Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg/Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg/Saar, Germany
| |
Collapse
|
37
|
Cucchiarini M, Henrionnet C, Mainard D, Pinzano A, Madry H. New trends in articular cartilage repair. J Exp Orthop 2015; 2:8. [PMID: 26914876 PMCID: PMC4544617 DOI: 10.1186/s40634-015-0026-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/25/2015] [Indexed: 12/31/2022] Open
Abstract
Damage to the articular cartilage is an important, prevalent, and unsolved clinical issue for the orthopaedic surgeon. This review summarizes innovative basic research approaches that may improve the current understanding of cartilage repair processes and lead to novel therapeutic options. In this regard, new aspects of cartilage tissue engineering with a focus on the choice of the best-suited cell source are presented. The importance of non-destructive cartilage imaging is highlighted with the recent availability of adapted experimental tools such as Second Harmonic Generation (SHG) imaging. Novel insights into cartilage pathophysiology based on the involvement of the infrapatellar fat pad in osteoarthritis are also described. Also, recombinant adeno-associated viral vectors are discussed as clinically adapted, efficient tools for potential gene-based medicines in a variety of articular cartilage disorders. Taken as a whole, such advances in basic research in diverse fields of articular cartilage repair may lead to the development of improved therapies in the clinics for an improved, effective treatment of cartilage lesions in a close future.
Collapse
Affiliation(s)
- Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center and Saarland University, Bldg 37, Kirrbergerstr. 1, D-66421, Homburg, Germany. .,Cartilage Net of the Greater Region, Homburg, Germany.
| | - Christel Henrionnet
- Cartilage Net of the Greater Region, Homburg, Germany. .,Ingénierie Moléculaire et Physiopathologie Articulaire, Unité Mixte de Recherches 7365 Centre National de la Recherche Scientifique, Université de Lorraine, F-54505, Vandoeuvre Lès Nancy, France.
| | - Didier Mainard
- Cartilage Net of the Greater Region, Homburg, Germany. .,Ingénierie Moléculaire et Physiopathologie Articulaire, Unité Mixte de Recherches 7365 Centre National de la Recherche Scientifique, Université de Lorraine, F-54505, Vandoeuvre Lès Nancy, France.
| | - Astrid Pinzano
- Cartilage Net of the Greater Region, Homburg, Germany. .,Ingénierie Moléculaire et Physiopathologie Articulaire, Unité Mixte de Recherches 7365 Centre National de la Recherche Scientifique, Université de Lorraine, F-54505, Vandoeuvre Lès Nancy, France.
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center and Saarland University, Bldg 37, Kirrbergerstr. 1, D-66421, Homburg, Germany. .,Cartilage Net of the Greater Region, Homburg, Germany. .,Department of Orthopaedic Surgery, Saarland University Medical Center and Saarland University, D-66421, Homburg/Saar, Germany.
| |
Collapse
|
38
|
Stoddart MJ, Bara J, Alini M. Cells and secretome--towards endogenous cell re-activation for cartilage repair. Adv Drug Deliv Rev 2015; 84:135-45. [PMID: 25174306 DOI: 10.1016/j.addr.2014.08.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 06/26/2014] [Accepted: 08/20/2014] [Indexed: 01/01/2023]
Abstract
Regenerative medicine approaches to cartilage tissue repair have mainly been concerned with the implantation of a scaffold material containing monolayer expanded cells into the defect, with the aim to differentiate the cells into chondrocytes. While this may be a valid approach, the secretome of the implanted cells and its effects on the endogenous resident cells, is gaining in interest. This review aims to summarize the knowledge on the secretome of mesenchymal stem cells, including knowledge from other tissues, in order to indicate how these mechanisms may be of value in repairing articular cartilage defects. Potential therapies and their effects on the repair of articular cartilage defects will be discussed, with a focus on the transition from classical cell therapy to the implantation of cell free matrices releasing specific cytokines.
Collapse
|
39
|
Elmallah RK, Cherian JJ, Jauregui JJ, Pierce TP, Beaver WB, Mont MA. Genetically modified chondrocytes expressing TGF-β1: a revolutionary treatment for articular cartilage damage? Expert Opin Biol Ther 2015; 15:455-64. [PMID: 25645308 DOI: 10.1517/14712598.2015.1009886] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Currently, joint arthroplasty remains the only definitive management of osteoarthritis, while other treatment modalities only provide temporary and symptomatic relief. The use of genetically engineered chondrocytes is currently undergoing clinical trials. Specifically, it has been designed to induce cartilage growth and differentiation in patients with degenerative arthritis, with the aim to play a curative role in the disease process. AREAS COVERED This treatment involves the incorporation of TGF-β1, which has been determined to play an influential role in chondrogenesis and extracellular matrix synthesis. Using genetic manipulation and viral transduction, TGF-β1 is incorporated into human chondrocytes and administered in a minimally invasive fashion directly to the affected joint. Following a database literature search, we evaluated the current evidence on this product and its outcomes. Furthermore, we also briefly reviewed other treatments developed for chondrogenesis and cartilage regeneration for comparison. EXPERT OPINION This treatment method has sustained positive effects on functional outcomes and cartilage growth in initial trials. It allows administration in a minimally invasive manner that does not require extended recovery time. Although several treatment modalities are currently under investigation and appear promising, we hope that these effects can be sustained in further studies. Ultimately, we anticipate that the results may be reproducible in many clinical settings and allow us to effectively treat cartilage damage in patients with degenerative arthritis.
Collapse
Affiliation(s)
- Randa K Elmallah
- Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, Center for Joint Preservation and Replacement , 2401 West Belvedere Avenue, Baltimore, MD 21215 , USA +1 410 601 8500 ; +1 410 601 8501 ; ;
| | | | | | | | | | | |
Collapse
|
40
|
Transforming growth factor Beta family: insight into the role of growth factors in regulation of fracture healing biology and potential clinical applications. Mediators Inflamm 2015; 2015:137823. [PMID: 25709154 PMCID: PMC4325469 DOI: 10.1155/2015/137823] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 11/09/2014] [Indexed: 01/15/2023] Open
Abstract
The transforming growth factor beta (TGF-β) family forms a group of three isoforms, TGF-β1, TGF-β2, and TGF-β3, with their structure formed by interrelated dimeric polypeptide chains. Pleiotropic and redundant functions of the TGF-β family concern control of numerous aspects and effects of cell functions, including proliferation, differentiation, and migration, in all tissues of the human body. Amongst many cytokines and growth factors, the TGF-β family is considered a group playing one of numerous key roles in control of physiological phenomena concerning maintenance of metabolic homeostasis in the bone tissue. By breaking the continuity of bone tissue, a spread-over-time and complex bone healing process is initiated, considered a recapitulation of embryonic intracartilaginous ossification. This process is a cascade of local and systemic phenomena spread over time, involving whole cell lineages and various cytokines and growth factors. Numerous in vivo and in vitro studies in various models analysing cytokines and growth factors' involvement have shown that TGF-β has a leading role in the fracture healing process. This paper sums up current knowledge on the basis of available literature concerning the role of the TGF-β family in the fracture healing process.
Collapse
|
41
|
Rey-Rico A, Frisch J, Venkatesan JK, Schmitt G, Madry H, Cucchiarini M. Determination of effective rAAV-mediated gene transfer conditions to support chondrogenic differentiation processes in human primary bone marrow aspirates. Gene Ther 2015; 22:50-7. [PMID: 25338919 DOI: 10.1038/gt.2014.90] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 07/23/2014] [Accepted: 08/26/2014] [Indexed: 11/09/2022]
Abstract
The genetic modification of freshly aspirated bone marrow may provide convenient tools to enhance the regenerative capacities of cartilage defects compared with the complex manipulation of isolated progenitor cells. In the present study, we examined the ability and safety of recombinant adeno-associated virus (rAAV) serotype 2 vectors to deliver various reporter gene sequences in primary human bone marrow aspirates over time without altering the chondrogenic processes in the samples. The results demonstrate that successful rAAV-mediated gene transfer and expression of the lacZ and red fluorescent protein marker genes were achieved in transduced aspirates at very high efficiencies (90-94%) and over extended periods of time (up to 125 days) upon treatment with hirudin, an alternative anticoagulant that does not prevent the adsorption of the rAAV-2 particles at the surface of their targets compared with heparin. Application of rAAV was safe, displaying neither cytotoxic nor detrimental effects on the cellular and proliferative activities or on the chondrogenic processes in the aspirates especially using an optimal dose of 0.5 mg ml(-1) hirudin, and application of the potent SOX9 transcription factor even enhanced these processes while counteracting hypertrophic differentiation. The current findings demonstrate the clinical value of this class of vector to durably and safely modify bone marrow aspirates as a means to further develop convenient therapeutic approaches to improve the healing of cartilage defects.
Collapse
Affiliation(s)
- A Rey-Rico
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - J Frisch
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - J K Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - G Schmitt
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - H Madry
- 1] Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany [2] Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg/Saar, Germany
| | - M Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| |
Collapse
|
42
|
Frisch J, Venkatesan JK, Rey-Rico A, Schmitt G, Madry H, Cucchiarini M. Determination of the Chondrogenic Differentiation Processes in Human Bone Marrow-Derived Mesenchymal Stem Cells Genetically Modified to Overexpress Transforming Growth Factor-β via Recombinant Adeno-Associated Viral Vectors. Hum Gene Ther 2014; 25:1050-60. [DOI: 10.1089/hum.2014.091] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- Janina Frisch
- Center of Experimental Orthopedics, Saarland University Medical Center, D-66421 Homburg, Germany
| | | | - Ana Rey-Rico
- Center of Experimental Orthopedics, Saarland University Medical Center, D-66421 Homburg, Germany
| | - Gertrud Schmitt
- Center of Experimental Orthopedics, Saarland University Medical Center, D-66421 Homburg, Germany
| | - Henning Madry
- Center of Experimental Orthopedics, Saarland University Medical Center, D-66421 Homburg, Germany
- Department of Orthopedic Surgery, Saarland University Medical Center, D-66421 Homburg, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopedics, Saarland University Medical Center, D-66421 Homburg, Germany
| |
Collapse
|
43
|
Moazedi-Fuerst FC, Hofner M, Gruber G, Weinhaeusel A, Stradner MH, Angerer H, Peischler D, Lohberger B, Glehr M, Leithner A, Sonntagbauer M, Graninger WB. Epigenetic differences in human cartilage between mild and severe OA. J Orthop Res 2014; 32:1636-45. [PMID: 25212754 DOI: 10.1002/jor.22722] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 07/24/2014] [Indexed: 02/04/2023]
Abstract
The development of osteoarthritis (OA) depends on genetic and environmental factors, which influence the biology of the chondrocyte via epigenetic regulation. Changes within the epigenome might lead the way to discovery of new pathogenetic pathways. We performed a genome-wide methylation screening to identify potential differences between paired mild and severe osteoarthritic human cartilage. Sixteen female patients suffering from OA underwent total knee joint replacement. Cartilage specimens collected from corresponding macroscopically undamaged and from damaged areas were processed for DNA extraction and histology to evaluate the histological grading of the disease. Paired specimens were analysed for the methylation status of the whole genome using human promoter microarrays (Agilent, Santa Clara, CA). Selected target genes were then validated via methylation-specific qPCR. One thousand two hundred and fourteen genetic targets were identified differentially methylated between mild and severe OA. One thousand and seventy of these targets were found hypermethylated and 144 hypomethylated. The descriptive analysis of these genes by Gene Ontology (GO), KEGG pathway and protein domain analyses points to pathways of development and differentiation. We identified a list of genes which are differently methylated in mild and severe OA cartilage. Within the pathways of growth and development new therapeutic targets might arise by improving our understanding of pathogenetic mechanisms in OA.
Collapse
|
44
|
Zhao R, Peng X, Li Q, Song W. Effects of phosphorylatable short peptide-conjugated chitosan-mediated IL-1Ra and igf-1 gene transfer on articular cartilage defects in rabbits. PLoS One 2014; 9:e112284. [PMID: 25390659 PMCID: PMC4229204 DOI: 10.1371/journal.pone.0112284] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 10/10/2014] [Indexed: 11/25/2022] Open
Abstract
Previously, we reported an improvement in the transfection efficiency of the plasmid DNA-chitosan (pDNA/CS) complex by the utilization of phosphorylatable short peptide-conjugated chitosan (pSP-CS). In this study, we investigated the effects of pSP-CS-mediated gene transfection of interleukin-1 receptor antagonist protein (IL-1Ra) combined with insulin-like growth factor-1 (IGF-1) in rabbit chondrocytes and in a rabbit model of cartilage defects. pBudCE4.1-IL-1Ra+igf-1, pBudCE4.1-IL-1Ra and pBudCE4.1-igf-1 were constructed and combined with pSP-CS to form pDNA/pSP-CS complexes. These complexes were transfected into rabbit primary chondrocytes or injected into the joint cavity. Seven weeks after treatment, all rabbits were sacrificed and analyzed. High levels of IL-1Ra and igf-1 expression were detected both in the cell culture supernatant and in the synovial fluid. In vitro, the transgenic complexes caused significant proliferation of chondrocytes, promotion of glycosaminoglycan (GAG) and collagen II synthesis, and inhibition of chondrocyte apoptosis and nitric oxide (NO) synthesis. In vivo, the exogenous genes resulted in increased collagen II synthesis and reduced NO and GAG concentrations in the synovial fluid; histological studies revealed that pDNA/pSP-CS treatment resulted in varying degrees of hyaline-like cartilage repair and Mankin score decrease. The co-expression of both genes produced greater effects than each single gene alone both in vitro and in vivo. The results suggest that pSP-CS is a good candidate for use in gene therapy for the treatment of cartilage defects and that igf-1 and IL-1Ra co-expression produces promising biologic effects on cartilage defects.
Collapse
Affiliation(s)
- Ronglan Zhao
- Department of Medical Laboratory, Shandong Provincial Key Laboratory of Clinical Laboratory Diagnostics, Weifang Medical University, Weifang, Shandong, China
| | - Xiaoxiang Peng
- Department of Medical Laboratory, Shandong Provincial Key Laboratory of Clinical Laboratory Diagnostics, Weifang Medical University, Weifang, Shandong, China
- * E-mail:
| | - Qian Li
- Department of Medical Laboratory, Shandong Provincial Key Laboratory of Clinical Laboratory Diagnostics, Weifang Medical University, Weifang, Shandong, China
| | - Wei Song
- Department of Medical Laboratory, Shandong Provincial Key Laboratory of Clinical Laboratory Diagnostics, Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|
45
|
Use of Tissue Engineering Strategies to Repair Joint Tissues in Osteoarthritis: Viral Gene Transfer Approaches. Curr Rheumatol Rep 2014; 16:449. [DOI: 10.1007/s11926-014-0449-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
46
|
Overexpression of human IGF-I via direct rAAV-mediated gene transfer improves the early repair of articular cartilage defects in vivo. Gene Ther 2014; 21:811-9. [PMID: 24989812 DOI: 10.1038/gt.2014.58] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 05/06/2014] [Accepted: 05/28/2014] [Indexed: 11/09/2022]
Abstract
Direct therapeutic gene transfer is a promising tool to treat articular cartilage defects. Here, we tested the ability of an recombinant adeno-associated virus (rAAV) insulin-like growth factor I (IGF-I) vector to improve the early repair of cartilage lesions in vivo. The vector was administered for 3 weeks in osteochondral defects created in the knee joints of rabbits compared with control (lacZ) treatment and in cells that participate in the repair processes (mesenchymal stem cells, chondrocytes). Efficient IGF-I expression was observed in the treated lesions and in isolated cells in vitro. rAAV-mediated IGF-I overexpression was capable of stimulating the biologic activities (proliferation, matrix synthesis) both in vitro and in vivo. IGF-I treatment in vivo was well tolerated, revealing significant improvements of the repair capabilities of the entire osteochondral unit. IGF-I overexpression delayed terminal differentiation and hypertrophy in the newly formed cartilage, possibly due to contrasting effects upon the osteogenic expression of RUNX2 and β-catenin and to stimulating effects of this factor on the parathyroid hormone/parathyroid hormone-related protein pathway in this area. Production of IGF-I improved the reconstitution of the subchondral bone layer in the defects, showing increased RUNX2 expression levels in this zone. These findings show the potential of directly applying therapeutic rAAVs to treat cartilage lesions.
Collapse
|
47
|
Gögebakan B, Uruc V, Ozden R, Duman IG, Yagiz AE, Okuyan HM, Aldemir O, Dogramaci Y, Kalaci A. Urotensin II (U-II), a novel cyclic peptide, possibly associated with the pathophysiology of osteoarthritis. Peptides 2014; 54:159-61. [PMID: 24468547 DOI: 10.1016/j.peptides.2014.01.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 01/16/2014] [Accepted: 01/16/2014] [Indexed: 01/27/2023]
Abstract
Synovial fibrosis is one of the main outcomes of osteoarthritis. Some authors have reported that urotensin-II (U-II) may cause pathologic fibrosis in cardiovascular system, lung and liver. However there are no previous reports available in the literature about its relationship with the synovial fibrosis in osteoarthritis. The aim of this study was to compare the U-II levels in knee synovial fluids obtained from osteoarthritic and non-osteoarthritic patients. Two groups were created, the osteoarthritis group and non-osteoarthritic control group. The control group was consisted of patients who underwent arthroscopic surgery for other reasons than cartilage disorders. In the osteoarthritis group all patients had grade 4 primer degenerative osteoarthritis and were treated with total knee arthroplasty. Minimum 1 mL knee synovial fluids were obtained during operation. Levels of U-II were measured by using ELISA kit U-II levels were significantly higher in the osteoarthritic group than that in the control group. No correlation was found between U-II levels and age. In conclusion, the significantly high U-II levels in the knee synovial fluid of osteoarthritic patients supported our hypothesis that "U-II may be associated with the synovial fibrosis in osteoarthritis".
Collapse
Affiliation(s)
- Bülent Gögebakan
- Mustafa Kemal University, Faculty of Medicine, Department of Medical Biology, 31034 Hatay, Turkey.
| | - Vedat Uruc
- Mustafa Kemal University, Faculty of Medicine, Department of Orthopedics and Traumatology, 31034 Hatay, Turkey
| | - Raif Ozden
- Mustafa Kemal University, Faculty of Medicine, Department of Orthopedics and Traumatology, 31034 Hatay, Turkey
| | - Ibrahim Gokhan Duman
- Mustafa Kemal University, Faculty of Medicine, Department of Orthopedics and Traumatology, 31034 Hatay, Turkey
| | - Abdullah Erman Yagiz
- Mustafa Kemal University, Faculty of Medicine, Department of Physical Therapy and Rehabilitation, 31034 Hatay, Turkey
| | - Hamza Malik Okuyan
- Mustafa Kemal University, Faculty of Medicine, Department of Medical Biology, 31034 Hatay, Turkey
| | - Ozgur Aldemir
- Mustafa Kemal University, Faculty of Medicine, Department of Medical Biology, 31034 Hatay, Turkey
| | - Yunus Dogramaci
- Mustafa Kemal University, Faculty of Medicine, Department of Orthopedics and Traumatology, 31034 Hatay, Turkey
| | - Aydiner Kalaci
- Mustafa Kemal University, Faculty of Medicine, Department of Orthopedics and Traumatology, 31034 Hatay, Turkey
| |
Collapse
|