1
|
Singh H, Dhanka M, Yadav I, Gautam S, Bashir SM, Mishra NC, Arora T, Hassan S. Technological Interventions Enhancing Curcumin Bioavailability in Wound-Healing Therapeutics. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:230-253. [PMID: 37897069 DOI: 10.1089/ten.teb.2023.0085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Abstract
Wound healing has been a challenge in the medical field. Tremendous research has been carried out to expedite wound healing by fabricating various formulations, some of which are now commercially available. However, owing to their natural source, people have been attracted to advanced formulations with herbal components. Among various herbs, curcumin has been the center of attraction from ancient times for its healing properties due to its multiple therapeutic effects, including antioxidant, antimicrobial, anti-inflammatory, anticarcinogenic, neuroprotective, and radioprotective properties. However, curcumin has a low water solubility and rapidly degrades into inactive metabolites, which limits its therapeutic efficacy. Henceforth, a carrier system is needed to carry curcumin, guard it against degradation, and keep its bioavailability and effectiveness. Different formulations with curcumin have been synthesized, and exist in the form of various synthetic and natural materials, including nanoparticles, hydrogels, scaffolds, films, fibers, and nanoemulgels, improving its bioavailability dramatically. This review discusses the advances in different types of curcumin-based formulations used in wound healing in recent times, concentrating on its mechanisms of action and discussing the updates on its application at several stages of the wound healing process. Impact statement Curcumin is a herbal compound extracted from turmeric root and has been used since time immemorial for its health benefits including wound healing. In clinical formulations, curcumin shows low bioavailability, which mainly stems from the way it is delivered in the body. Henceforth, a carrier system is needed to carry curcumin, guard it against degradation, while maintaining its bioavailability and therapeutic efficacy. This review offers an overview of the advanced technological interventions through tissue engineering approaches to efficiently utilize curcumin in different types of wound healing applications.
Collapse
Affiliation(s)
- Hemant Singh
- Department of Polymer and Process Engineering, Indian Institute of Technology Roorkee, Roorkee, India
- Department of Biological Engineering, Indian Institute of Technology Gandhinagar, Gandhinagar, India
- Department of Biology, Khalifa University, Main Campus, Abu Dhabi, United Arab Emirates
- Advanced Materials Chemistry Center, Khalifa University, SAN Campus, Abu Dhabi, United Arab Emirates
| | - Mukesh Dhanka
- Department of Biological Engineering, Indian Institute of Technology Gandhinagar, Gandhinagar, India
| | - Indu Yadav
- Department of Polymer and Process Engineering, Indian Institute of Technology Roorkee, Roorkee, India
| | - Sneh Gautam
- Department of Molecular Biology & Genetic Engineering, G. B. Pant University of Agriculture & Technology, Pantnagar, India
| | - Showkeen Muzamil Bashir
- Biochemistry and Molecular Biology Lab Division, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Srinagar, India
| | - Narayan Chandra Mishra
- Department of Polymer and Process Engineering, Indian Institute of Technology Roorkee, Roorkee, India
| | - Taruna Arora
- Reproductive Health Division of RBMCH, Indian Council of Medical Research, New Delhi, India
| | - Shabir Hassan
- Department of Biology, Khalifa University, Main Campus, Abu Dhabi, United Arab Emirates
- Advanced Materials Chemistry Center, Khalifa University, SAN Campus, Abu Dhabi, United Arab Emirates
| |
Collapse
|
2
|
Fan M, Qiang L, Wang Y, Liu Y, Zhuang H, Guo R, Ben Y, Li Q, Zheng P. 3D bioprinted hydrogel/polymer scaffold with factor delivery and mechanical support for growth plate injury repair. Front Bioeng Biotechnol 2023; 11:1210786. [PMID: 37324424 PMCID: PMC10265638 DOI: 10.3389/fbioe.2023.1210786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 05/23/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction: Growth plate injury is a significant challenge in clinical practice, as it could severely affect the limb development of children, leading to limb deformity. Tissue engineering and 3D bioprinting technology have great potential in the repair and regeneration of injured growth plate, but there are still challenges associated with achieving successful repair outcomes. Methods: In this study, GelMA hydrogel containing PLGA microspheres loaded with chondrogenic factor PTH(1-34) was combined with BMSCs and Polycaprolactone (PCL) to develop the PTH(1-34)@PLGA/BMSCs/GelMA-PCL scaffold using bio-3D printing technology. Results: The scaffold exhibited a three-dimensional interconnected porous network structure, good mechanical properties, biocompatibility, and was suitable for cellchondrogenic differentiation. And a rabbit model of growth plate injury was appliedto validate the effect of scaffold on the repair of injured growth plate. The resultsshowed that the scaffold was more effective than injectable hydrogel in promotingcartilage regeneration and reducing bone bridge formation. Moreover, the addition ofPCL to the scaffold provided good mechanical support, significantly reducing limbdeformities after growth plate injury compared with directly injected hydrogel. Discussion: Accordingly, our study demonstrates the feasibility of using 3D printed scaffolds for treating growth plate injuries and could offer a new strategy for the development of growth plate tissue engineering therapy.
Collapse
Affiliation(s)
- Minjie Fan
- Department of Orthopaedic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Qiang
- Department of Orthopaedic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Advanced Technologies of Materials (MOE), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Yiwei Wang
- Department of Orthopaedic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yihao Liu
- Department of Orthopaedic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hanjie Zhuang
- Department of Orthopaedic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ruoyi Guo
- Department of Orthopaedic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yulong Ben
- Department of Orthopaedic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qiang Li
- Department of Orthopaedic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Pengfei Zheng
- Department of Orthopaedic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
3
|
Weißenberger M, Wagenbrenner M, Nickel J, Ahlbrecht R, Blunk T, Steinert AF, Gilbert F. Comparative in vitro treatment of mesenchymal stromal cells with GDF-5 and R57A induces chondrogenic differentiation while limiting chondrogenic hypertrophy. J Exp Orthop 2023; 10:29. [PMID: 36943593 PMCID: PMC10030724 DOI: 10.1186/s40634-023-00594-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 03/08/2023] [Indexed: 03/23/2023] Open
Abstract
PURPOSE Hypertrophic cartilage is an important characteristic of osteoarthritis and can often be found in patients suffering from osteoarthritis. Although the exact pathomechanism remains poorly understood, hypertrophic de-differentiation of chondrocytes also poses a major challenge in the cell-based repair of hyaline cartilage using mesenchymal stromal cells (MSCs). While different members of the transforming growth factor beta (TGF-β) family have been shown to promote chondrogenesis in MSCs, the transition into a hypertrophic phenotype remains a problem. To further examine this topic we compared the effects of the transcription growth and differentiation factor 5 (GDF-5) and the mutant R57A on in vitro chondrogenesis in MSCs. METHODS Bone marrow-derived MSCs (BMSCs) were placed in pellet culture and in-cubated in chondrogenic differentiation medium containing R57A, GDF-5 and TGF-ß1 for 21 days. Chondrogenesis was examined histologically, immunohistochemically, through biochemical assays and by RT-qPCR regarding the expression of chondrogenic marker genes. RESULTS Treatment of BMSCs with R57A led to a dose dependent induction of chondrogenesis in BMSCs. Biochemical assays also showed an elevated glycosaminoglycan (GAG) content and expression of chondrogenic marker genes in corresponding pellets. While treatment with R57A led to superior chondrogenic differentiation compared to treatment with the GDF-5 wild type and similar levels compared to incubation with TGF-ß1, levels of chondrogenic hypertrophy were lower after induction with R57A and the GDF-5 wild type. CONCLUSIONS R57A is a stronger inducer of chondrogenesis in BMSCs than the GDF-5 wild type while leading to lower levels of chondrogenic hypertrophy in comparison with TGF-ß1.
Collapse
Affiliation(s)
- Manuel Weißenberger
- Department of Orthopaedic Surgery, Center for Musculoskeletal Research, Julius-Maximilians-University Würzburg, König-Ludwig-Haus, Würzburg, Germany.
- Department of Orthopedic Surgery, University of Wuerzburg, König-Ludwig-Haus, Brettreichstraße 11, 97074, Würzburg, Germany.
| | - Mike Wagenbrenner
- Department of Orthopaedic Surgery, Center for Musculoskeletal Research, Julius-Maximilians-University Würzburg, König-Ludwig-Haus, Würzburg, Germany
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - Joachim Nickel
- Department of Tissue Engineering and Regenerative Medicine, Julius-Maximilians-University Würzburg, University Hospital, Würzburg, Germany
| | - Rasmus Ahlbrecht
- Department of Orthopaedic Surgery, Center for Musculoskeletal Research, Julius-Maximilians-University Würzburg, König-Ludwig-Haus, Würzburg, Germany
- Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Julius-Maximilians-University Würzburg, University Hospital, Würzburg, Germany
| | - Torsten Blunk
- Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Julius-Maximilians-University Würzburg, University Hospital, Würzburg, Germany
| | - Andre F Steinert
- Department of Orthopaedic Surgery, Center for Musculoskeletal Research, Julius-Maximilians-University Würzburg, König-Ludwig-Haus, Würzburg, Germany
- Current address:, Department of Orthopaedic, Trauma, Shoulder and Arthroplasty Surgery, Rhön-Klinikum, Campus Bad Neustadt, Bad Neustadt, Germany
| | - Fabian Gilbert
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), University Hospital, LMU Munich, Marchioninistraße 15, 81377, Munich, Germany
- Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Julius-Maximilians-University Würzburg, University Hospital, Würzburg, Germany
| |
Collapse
|
4
|
Yergeshov AA, Zoughaib M, Ishkaeva RA, Savina IN, Abdullin TI. Regenerative Activities of ROS-Modulating Trace Metals in Subcutaneously Implanted Biodegradable Cryogel. Gels 2022; 8:118. [PMID: 35200498 PMCID: PMC8872170 DOI: 10.3390/gels8020118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 01/27/2023] Open
Abstract
Divalent trace metals (TM), especially copper (Cu), cobalt (Co) and zinc (Zn), are recognized as essential microelements for tissue homeostasis and regeneration. To achieve a balance between therapeutic activity and safety of administered TMs, effective gel formulations of TMs with elucidated regenerative mechanisms are required. We studied in vitro and in vivo effects of biodegradable macroporous cryogels doped with Cu, Co or Zn in a controllable manner. The extracellular ROS generation by metal dopants was assessed and compared with the intracellular effect of soluble TMs. The stimulating ability of TMs in the cryogels for cell proliferation, differentiation and cytokine/growth factor biosynthesis was characterized using HSF and HUVEC primary human cells. Multiple responses of host tissues to the TM-doped cryogels upon subcutaneous implantation were characterized taking into account the rate of biodegradation, production of HIF-1α/matrix metalloproteinases and the appearance of immune cells. Cu and Zn dopants did not disturb the intact skin organization while inducing specific stimulating effects on different skin structures, including vasculature, whereas Co dopant caused a significant reorganization of skin layers, the appearance of multinucleated giant cells, along with intense angiogenesis in the dermis. The results specify and compare the prooxidant and regenerative potential of Cu, Co and Zn-doped biodegradable cryogels and are of particular interest for the development of advanced bioinductive hydrogel materials for controlling angiogenesis and soft tissue growth.
Collapse
Affiliation(s)
- Abdulla A. Yergeshov
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia; (A.A.Y.); (M.Z.); (R.A.I.)
| | - Mohamed Zoughaib
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia; (A.A.Y.); (M.Z.); (R.A.I.)
| | - Rezeda A. Ishkaeva
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia; (A.A.Y.); (M.Z.); (R.A.I.)
| | - Irina N. Savina
- School of Applied Sciences, University of Brighton, Huxley Building, Lewes Road, Brighton BN2 4GJ, UK;
| | - Timur I. Abdullin
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia; (A.A.Y.); (M.Z.); (R.A.I.)
| |
Collapse
|
5
|
Iwan A, Moskalewski S, Hyc A. Growth factor profile in calcified cartilage from the metaphysis of a calf costochondral junction, the site of initial bone formation. Biomed Rep 2021; 14:54. [PMID: 33884197 PMCID: PMC8056382 DOI: 10.3892/br.2021.1430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 03/11/2021] [Indexed: 12/25/2022] Open
Abstract
Endochondral bone formation is orchestrated by growth factors produced by chondrocytes and deposited in the cartilage matrix. Whilst some of these factors have been identified, the complete list and their relationship remains unknown. In the present study, the growth factors were isolated from non-calcified and calcified cartilage of costochondral junctions. Cartilage dissected from the ribs of 6-20-week-old calves was purchased from a local butcher within 24 h of the death of the animal. The isolation involved hyaluronidase digestion, guanidinium hydrochloride (GuHCl) extraction, HCl decalcification and GuHCl extraction of the decalcified matrix. Growth factors were purified by heparin chromatography and their quantities were estimated using ELISA. Decalcified cartilage was also used for protein sequence analysis (data are available via ProteomeXchange; ID, PXD021781). Bone morphogenetic protein-7 (BMP-7), growth/differentiation factor-5 (GDF-5) and NEL-like protein-1 (NELL-1), all known growth factors that stimulate bone formation, quantitatively accounted for the majority of the material obtained in all steps of isolation. Thus, cartilage serves as a store for growth factors. During initial bone formation septoclasts release osteoclastogenesis-stimulating factors deposited in non-calcified cartilage. Osteoclasts dissolve calcified cartilage and transport the released factors required for the stimulation of osteoprogenitor cells to deposit osteoid. High concentrations of BMP-7, GDF-5 and NELL-1 at the site of initial bone formation may suggest that their synergistic action favours osteogenesis.
Collapse
Affiliation(s)
- Anna Iwan
- Department of Histology and Embryology, Medical University of Warsaw, Warsaw PL02004, Poland
| | - Stanisław Moskalewski
- Department of Histology and Embryology, Medical University of Warsaw, Warsaw PL02004, Poland
| | - Anna Hyc
- Department of Histology and Embryology, Medical University of Warsaw, Warsaw PL02004, Poland
| |
Collapse
|
6
|
Sheikh MA, Emerald BS, Ansari SA. Stem cell fate determination through protein O-GlcNAcylation. J Biol Chem 2021; 296:100035. [PMID: 33154167 PMCID: PMC7948975 DOI: 10.1074/jbc.rev120.014915] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 11/05/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022] Open
Abstract
Embryonic and adult stem cells possess the capability of self-renewal and lineage-specific differentiation. The intricate balance between self-renewal and differentiation is governed by developmental signals and cell-type-specific gene regulatory mechanisms. A perturbed intra/extracellular environment during lineage specification could affect stem cell fate decisions resulting in pathology. Growing evidence demonstrates that metabolic pathways govern epigenetic regulation of gene expression during stem cell fate commitment through the utilization of metabolic intermediates or end products of metabolic pathways as substrates for enzymatic histone/DNA modifications. UDP-GlcNAc is one such metabolite that acts as a substrate for enzymatic mono-glycosylation of various nuclear, cytosolic, and mitochondrial proteins on serine/threonine amino acid residues, a process termed protein O-GlcNAcylation. The levels of GlcNAc inside the cells depend on the nutrient availability, especially glucose. Thus, this metabolic sensor could modulate gene expression through O-GlcNAc modification of histones or other proteins in response to metabolic fluctuations. Herein, we review evidence demonstrating how stem cells couple metabolic inputs to gene regulatory pathways through O-GlcNAc-mediated epigenetic/transcriptional regulatory mechanisms to govern self-renewal and lineage-specific differentiation programs. This review will serve as a primer for researchers seeking to better understand how O-GlcNAc influences stemness and may catalyze the discovery of new stem-cell-based therapeutic approaches.
Collapse
Affiliation(s)
- Muhammad Abid Sheikh
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE; Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE
| | - Suraiya Anjum Ansari
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE; Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE.
| |
Collapse
|
7
|
Al-Masawa ME, Wan Kamarul Zaman WS, Chua KH. Biosafety evaluation of culture-expanded human chondrocytes with growth factor cocktail: a preclinical study. Sci Rep 2020; 10:21583. [PMID: 33299022 PMCID: PMC7725787 DOI: 10.1038/s41598-020-78395-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/27/2020] [Indexed: 01/03/2023] Open
Abstract
The scarcity of chondrocytes is a major challenge for cartilage tissue engineering. Monolayer expansion is necessary to amplify the limited number of chondrocytes needed for clinical application. Growth factors are often added to improve monolayer culture conditions, promoting proliferation, and enhancing chondrogenesis. Limited knowledge on the biosafety of the cell products manipulated with growth factors in culture has driven this study to evaluate the impact of growth factor cocktail supplements in chondrocyte culture medium on chondrocyte genetic stability and tumorigenicity. The growth factors were basic fibroblast growth factor (b-FGF), transforming growth factor β2 (TGF β2), insulin-like growth factor 1 (IGF-1), insulin-transferrin-selenium (ITS), and platelet-derived growth factor (PD-GF). Nasal septal chondrocytes cultured in growth factor cocktail exhibited a significantly high proliferative capacity. Comet assay revealed no significant DNA damage. Flow cytometry showed chondrocytes were mostly at G0-G1 phase, exhibiting normal cell cycle profile with no aneuploidy. We observed a decreased tumour suppressor genes’ expression (p53, p21, pRB) and no TP53 mutations or tumour formation after 6 months of implantation in nude mice. Our data suggest growth factor cocktail has a low risk of inducing genotoxic and tumorigenic effects on chondrocytes up to passage 6 with 16.6 population doublings. This preclinical tumorigenicity and genetic instability evaluation is crucial for further clinical works.
Collapse
Affiliation(s)
- Maimonah-Eissa Al-Masawa
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, 56000, Kuala Lumpur, Malaysia.
| | | | - Kien-Hui Chua
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, 56000, Kuala Lumpur, Malaysia.
| |
Collapse
|
8
|
Barlian A, Judawisastra H, Alfarafisa NM, Wibowo UA, Rosadi I. Chondrogenic differentiation of adipose-derived mesenchymal stem cells induced by L-ascorbic acid and platelet rich plasma on silk fibroin scaffold. PeerJ 2018; 6:e5809. [PMID: 30488014 PMCID: PMC6250097 DOI: 10.7717/peerj.5809] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 09/21/2018] [Indexed: 12/22/2022] Open
Abstract
Articular cartilage is an avascular tissue with limited regenerative property. Therefore, a defect or trauma in articular cartilage due to disease or accident can lead to progressive tissue deterioration. Cartilage tissue engineering, by replacing defective cartilage tissue, is a method for repairing such a problem. In this research, three main aspects-cell, biomaterial scaffold, and bioactive factors-that support tissue engineering study were optimized. Adipose-derived mesenchymal stem cells (ADSC) that become cartilage were grown in an optimized growth medium supplemented with either platelet rich plasma (PRP) or L-ascorbic acid (LAA). As the characterization result, the ADSC used in this experiment could be classified as Mesenchymal Stem Cell (MSC) based on multipotency analysis and cell surface marker analysis. The biomaterial scaffold was fabricated from the Bombyx morii cocoon using silk fibroin by salt leaching method and was engineered to form different sizes of pores to provide optimized support for cell adhesion and growth. Biocompatibility and cytotoxicity evaluation was done using MTT assay to optimize silk fibroin concentration and pore size. Characterized ADSC were grown on the optimized scaffold. LAA and PRP were chosen as bioactive factors to induce ADSC differentiation to become chondrocytes. The concentration optimization of LAA and PRP was analyzed by cell proliferation using MTT assay and chondrogenic differentiation by measuring glycosaminoglycan (GAG) using Alcian Blue at 605 nm wavelength. The optimum silk fibroin concentration, pore size, LAA concentration, and PRP concentration were used to grow and differentiate characterized ADSC for 7, 14, and 21 days. The cell morphology on the scaffold was analyzed using a scanning electron microscope (SEM). The result showed that the ADSC could adhere on plastic, express specific cell surface markers (CD73, CD90, and CD105), and could be differentiated into three types of mature cells. The silk fibroin scaffold made from 12% w/v concentration formed a 500 µm pore diameter (SEM analysis), and was shown by MTT assay to be biocompatible and to facilitate cell growth. The optimum concentrations of the bioactive factors LAA and PRP were 50 µg/mL and 10%, respectively. GAG analysis with Alcian Blue staining suggested that PRP induction medium and LAA induction medium on 12% w/v scaffold could effectively promote not only cell adhesion and cell proliferation but also chondrogenic differentiation of ADSC within 21 days of culture. Therefore, this study provides a new approach to articular tissue engineering with a combination of ADSC as cell source, LAA and PRP as bioactive factors, and silk fibroin as a biocompatible and biodegradable scaffold.
Collapse
Affiliation(s)
- Anggraini Barlian
- School of Life Sciences and Technology, Institute of Technology Bandung, Bandung, West Java, Indonesia
| | - Hermawan Judawisastra
- Faculty of Mechanical and Aerospace Engineering, Institute of Technology Bandung, Bandung, West Java, Indonesia
| | - Nayla M Alfarafisa
- School of Life Sciences and Technology, Institute of Technology Bandung, Bandung, West Java, Indonesia
| | - Untung A Wibowo
- Faculty of Mechanical and Aerospace Engineering, Institute of Technology Bandung, Bandung, West Java, Indonesia
| | | |
Collapse
|
9
|
Different Polyubiquitinated Bodies in Human Dendritic Cells: IL-4 Causes PaCS During Differentiation while LPS or IFNα Induces DALIS During Maturation. Sci Rep 2017; 7:1844. [PMID: 28500302 PMCID: PMC5431999 DOI: 10.1038/s41598-017-02090-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 04/05/2017] [Indexed: 01/08/2023] Open
Abstract
Two types of polyubiquitin-reactive cytoplasmic bodies, particulate cytoplasmic structures (PaCS) and dendritic cell (DC) aggresome-like induced structures (DALIS), were analyzed by electron microscopy, immunocytochemistry, immunoblotting, and flow cytometry in DC obtained from human blood monocytes incubated with GM-CSF plus IL-4 (IL4-DC), GM-CSF plus IFNα (IFN-DC), or GM-CSF alone (GM-DC), with or without LPS maturation. PaCS developed as monomorphic aggregates of proteasome-reactive barrel-like particles only in ribosomes-rich cytoplasmic areas of differentiating IL4-DC. In contrast, DALIS formed as vesicular bodies storing K63-linked ubiquitinated proteins by coalescence of increased endosomal structures, in IFN-DC or after LPS maturation of GM-DC. DALIS-forming cells showed incomplete morphological and functional DC-type differentiation when compared to PaCS-forming IL4-DC. PaCS and DALIS may have different function as well as different origin and cytochemistry. DALIS may be a transient accumulation site of potentially antigenic polyubiquitinated proteins during their processing and presentation. PaCS are found under physiologic or pathologic conditions associated with increased/deranged protein synthesis and increased ubiquitin-proteasome activity. Given its high heat-shock protein content PaCS may work as a quality control structure for newly synthesized, cytosolic proteins. This comparative analysis suggests that PaCS and DALIS have distinctive roles in DC.
Collapse
|
10
|
Hemphill DD, McIlwraith CW, Slayden RA, Samulski RJ, Goodrich LR. Adeno-associated virus gene therapy vector scAAVIGF-I for transduction of equine articular chondrocytes and RNA-seq analysis. Osteoarthritis Cartilage 2016; 24:902-11. [PMID: 26706703 DOI: 10.1016/j.joca.2015.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 11/08/2015] [Accepted: 12/01/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE IGF-I is one of several anabolic factors being investigated for the treatment of osteoarthritis (OA). Due to the short biological half-life, extended administration is required for more robust cartilage healing. Here we create a self-complimentary adeno-associated virus (AAV) gene therapy vector utilizing the transgene for IGF-I. DESIGN Various biochemical assays were performed to investigate the cellular response to scAAVIGF-I treatment vs an scAAVGFP positive transduction control and a negative for transduction control culture. RNA-sequencing analysis was also performed to establish a differential regulation profile of scAAVIGF-I transduced chondrocytes. RESULTS Biochemical analyses indicated an average media IGF-I concentration of 608 ng/ml in the scAAVIGF-I transduced chondrocytes. This increase in IGF-I led to increased expression of collagen type II and aggrecan and increased protein concentrations of cellular collagen type II and media glycosaminoglycan vs both controls. RNA-seq revealed a global regulatory pattern consisting of 113 differentially regulated GO categories including those for chondrocyte and cartilage development and regulation of apoptosis. CONCLUSIONS This research substantiates that scAAVIGF-I gene therapy vector increased production of IGF-I to clinically relevant levels with a biological response by chondrocytes conducive to increased cartilage healing. The RNA-seq further established a set of differentially expressed genes and gene ontologies induced by the scAAVIGF-I vector while controlling for AAV infection. This dataset provides a static representation of the cellular transcriptome that, while only consisting of one time point, will allow for further gene expression analyses to compare additional cartilage healing therapeutics or a transient cellular response.
Collapse
Affiliation(s)
- D D Hemphill
- Orthopaedic Research Center, College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA.
| | - C W McIlwraith
- Orthopaedic Research Center, College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA.
| | - R A Slayden
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA.
| | - R J Samulski
- University of North Carolina Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - L R Goodrich
- Orthopaedic Research Center, College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
11
|
Jaroszewicz J, Kosowska A, Hutmacher D, Swieszkowski W, Moskalewski S. Insight into characteristic features of cartilage growth plate as a physiological template for bone formation. J Biomed Mater Res A 2015; 104:357-66. [DOI: 10.1002/jbm.a.35575] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 09/08/2015] [Accepted: 09/23/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Jakub Jaroszewicz
- Faculty of Materials Science and Engineering; Warsaw University of Technology; Woloska 141 Warsaw 02-507 Poland
| | - Anna Kosowska
- Department of Histology and Embryology; Medical University of Warsaw; Chalubinskiego 5 Warsaw 02-004 Poland
| | - Dietmar Hutmacher
- Institute of Health and Biomedical Innovation, Queensland University of Technology; 60 Musk Avenue Kelvin Grove QLD 4059 Australia
| | - Wojciech Swieszkowski
- Faculty of Materials Science and Engineering; Warsaw University of Technology; Woloska 141 Warsaw 02-507 Poland
| | - Stanisław Moskalewski
- Department of Histology and Embryology; Medical University of Warsaw; Chalubinskiego 5 Warsaw 02-004 Poland
| |
Collapse
|
12
|
Chung CY, Heebner J, Baskaran H, Welter JF, Mansour JM. Ultrasound Elastography for Estimation of Regional Strain of Multilayered Hydrogels and Tissue-Engineered Cartilage. Ann Biomed Eng 2015; 43:2991-3003. [PMID: 26077987 DOI: 10.1007/s10439-015-1356-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 06/05/2015] [Indexed: 01/07/2023]
Abstract
Tissue-engineered (TE) cartilage constructs tend to develop inhomogeneously, thus, to predict the mechanical performance of the tissue, conventional biomechanical testing, which yields average material properties, is of limited value. Rather, techniques for evaluating regional and depth-dependent properties of TE cartilage, preferably non-destructively, are required. The purpose of this study was to build upon our previous results and to investigate the feasibility of using ultrasound elastography to non-destructively assess the depth-dependent biomechanical characteristics of TE cartilage while in a sterile bioreactor. As a proof-of-concept, and to standardize an assessment protocol, a well-characterized three-layered hydrogel construct was used as a surrogate for TE cartilage, and was studied under controlled incremental compressions. The strain field of the construct predicted by elastography was then validated by comparison with a poroelastic finite-element analysis (FEA). On average, the differences between the strains predicted by elastography and the FEA were within 10%. Subsequently engineered cartilage tissue was evaluated in the same test fixture. Results from these examinations showed internal regions where the local strain was 1-2 orders of magnitude greater than that near the surface. These studies document the feasibility of using ultrasound to evaluate the mechanical behaviors of maturing TE constructs in a sterile environment.
Collapse
Affiliation(s)
- Chen-Yuan Chung
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, 2123 Martin Luther King Jr. Drive, Glennan Building Room 616A, Cleveland, OH, 44106, USA.,Department of Mechanical Engineering, National Central University, Taoyuan, Taiwan
| | - Joseph Heebner
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, 2123 Martin Luther King Jr. Drive, Glennan Building Room 616A, Cleveland, OH, 44106, USA
| | - Harihara Baskaran
- Department of Chemical and Biomolecular Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Jean F Welter
- Department of Biology (Skeletal Research Center), Case Western Reserve University, Cleveland, OH, USA
| | - Joseph M Mansour
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, 2123 Martin Luther King Jr. Drive, Glennan Building Room 616A, Cleveland, OH, 44106, USA.
| |
Collapse
|
13
|
De Leonardis F, Monti L, Gualeni B, Tenni R, Forlino A, Rossi A. Altered signaling in the G1 phase deregulates chondrocyte growth in a mouse model with proteoglycan undersulfation. J Cell Biochem 2015; 115:1779-86. [PMID: 24820054 PMCID: PMC4262066 DOI: 10.1002/jcb.24844] [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] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 05/09/2014] [Indexed: 12/15/2022]
Abstract
In several skeletal dysplasias defects in extracellular matrix molecules affect not only the structural and mechanical properties of cartilage, but also the complex network of signaling pathways involved in cell proliferation and differentiation. Sulfated proteoglycans, besides playing an important structural role in cartilage, are crucial in modulating the transport, diffusion, and interactions of growth factors with their specific targets, taking part in the regulation of signaling pathways involved in skeletal development and growth. In this work, we investigated by real time PCR and Western blots of the microdissected growth plate and by immunohistochemistry the molecular basis of reduced chondrocyte proliferation in the growth plate of the dtd mouse, a chondrodysplastic model with defective chondroitin sulfate proteoglycan sulfation of articular and growth plate cartilage. We detected activation of the Wnt pathway, leading to an increase in the non-phosphorylated form of nuclear β-catenin and subsequent up-regulation of cyclin D1 expression in the G1 phase of the cell cycle. β-Catenin was further stabilized by up-regulation of Smad3 expression through TGF-β pathway synergistic activation. We demonstrate that notwithstanding cyclin D1 expression increase, cell cycle progression is compromised in the G1 phase due to reduced phosphorylation of the pocket protein p130 leading to inhibition of transcription factors of the E2F family which are crucial for cell cycle progression and DNA replication. These data, together with altered Indian hedgehox signaling detected previously, explain at the molecular level the reduced chondrocyte proliferation rate of the dtd growth plate leading to reduced skeletal growth. J. Cell. Biochem. 115: 1779–1786, 2014.
Collapse
Affiliation(s)
- Fabio De Leonardis
- Department of Molecular Medicine, Unit of Biochemistry, University of Pavia, Pavia, Italy
| | | | | | | | | | | |
Collapse
|
14
|
Lam J, Lu S, Kasper FK, Mikos AG. Strategies for controlled delivery of biologics for cartilage repair. Adv Drug Deliv Rev 2015; 84:123-34. [PMID: 24993610 DOI: 10.1016/j.addr.2014.06.006] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 04/28/2014] [Accepted: 06/24/2014] [Indexed: 01/08/2023]
Abstract
The delivery of biologics is an important component in the treatment of osteoarthritis and the functional restoration of articular cartilage. Numerous factors have been implicated in the cartilage repair process, but the uncontrolled delivery of these factors may not only reduce their full reparative potential but can also cause unwanted morphological effects. It is therefore imperative to consider the type of biologic to be delivered, the method of delivery, and the temporal as well as spatial presentation of the biologic to achieve the desired effect in cartilage repair. Additionally, the delivery of a single factor may not be sufficient in guiding neo-tissue formation, motivating recent research toward the delivery of multiple factors. This review will discuss the roles of various biologics involved in cartilage repair and the different methods of delivery for appropriate healing responses. A number of spatiotemporal strategies will then be emphasized for the controlled delivery of single and multiple bioactive factors in both in vitro and in vivo cartilage tissue engineering applications.
Collapse
Affiliation(s)
- Johnny Lam
- Department of Bioengineering, Rice University, Houston, TX, United States
| | - Steven Lu
- Department of Bioengineering, Rice University, Houston, TX, United States
| | - F Kurtis Kasper
- Department of Bioengineering, Rice University, Houston, TX, United States
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX, United States; Department of Chemical and Biomolecular Engineering, Rice University, Houston, TX, United States.
| |
Collapse
|
15
|
|
16
|
Bach FC, Rutten K, Hendriks K, Riemers FM, Cornelissen P, de Bruin A, Arkesteijn GJ, Wubbolts R, Horton WA, Penning LC, Tryfonidou MA. The paracrine feedback loop between vitamin D₃ (1,25(OH)₂D₃) and PTHrP in prehypertrophic chondrocytes. J Cell Physiol 2014; 229:1999-2014. [PMID: 24777663 PMCID: PMC4298802 DOI: 10.1002/jcp.24658] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 04/25/2014] [Indexed: 12/16/2022]
Abstract
The endocrine feedback loop between vitamin D3 (1,25(OH)2D3) and parathyroid hormone (PTH) plays a central role in skeletal development. PTH-related protein (PTHrP) shares homology and its receptor (PTHR1) with PTH. The aim of this study was to investigate whether there is a functional paracrine feedback loop between 1,25(OH)2D3 and PTHrP in the growth plate, in parallel with the endocrine feedback loop between 1,25(OH)2D3 and PTH. This was investigated in ATDC5 cells treated with 10−8 M 1,25(OH)2D3 or PTHrP, Col2-pd2EGFP transgenic mice, and primary Col2-pd2EGFP growth plate chondrocytes isolated by FACS, using RT-qPCR, Western blot, PTHrP ELISA, chromatin immunoprecipitation (ChIP) assay, silencing of the 1,25(OH)2D3 receptor (VDR), immunofluorescent staining, immunohistochemistry, and histomorphometric analysis of the growth plate. The ChIP assay confirmed functional binding of the VDR to the PTHrP promoter, but not to the PTHR1 promoter. Treatment with 1,25(OH)2D3 decreased PTHrP protein production, an effect which was prevented by silencing of the VDR. Treatment with PTHrP significantly induced VDR production, but did not affect 1α- and 24-hydroxylase expression. Hypertrophic differentiation was inhibited by PTHrP and 1,25(OH)2D3 treatment. Taken together, these findings indicate that there is a functional paracrine feedback loop between 1,25(OH)2D3 and PTHrP in the growth plate. 1,25(OH)2D3 decreases PTHrP production, while PTHrP increases chondrocyte sensitivity to 1,25(OH)2D3 by increasing VDR production. In light of the role of 1,25(OH)2D3 and PTHrP in modulating chondrocyte differentiation, 1,25(OH)2D3 in addition to PTHrP could potentially be used to prevent undesirable hypertrophic chondrocyte differentiation during cartilage repair or regeneration.
Collapse
Affiliation(s)
- Frances C Bach
- Faculty of Veterinary Medicine, Department of Clinical Sciences of Companion Animals, Utrecht University, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Bae JW, Choi JH, Lee Y, Park KD. Horseradish peroxidase-catalysed in situ-forming hydrogels for tissue-engineering applications. J Tissue Eng Regen Med 2014; 9:1225-32. [PMID: 24916126 DOI: 10.1002/term.1917] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 01/26/2014] [Accepted: 04/22/2014] [Indexed: 12/18/2022]
Abstract
In situ-forming hydrogels are an attractive class of implantable biomaterials that are used for biomedical applications. These injectable hydrogels are versatile and provide a convenient platform for delivering cells and drugs via minimally invasive surgery. Although several crosslinking methods for preparing in situ forming hydrogels have been developed over the past two decades, most hydrogels are not sufficiently versatile for use in a wide variety of tissue-engineering applications. In recent years, enzyme-catalysed crosslinking approaches have been emerged as a new approach for developing in situ-forming hydrogels. In particular, the horseradish peroxidase (HRP)-catalysed crosslinking approach has received increasing interest, due to its highly improved and tunable capacity to obtain hydrogels with desirable properties. The HRP-catalysed crosslinking reaction immediately occurs upon mixing phenol-rich polymers with HRP and hydrogen peroxide (H2O2) in aqueous media. Based on this unique gel-forming feature, recent studies have shown that various properties of formed hydrogels, such as gelation time, stiffness and degradation rate, can be easily manipulated by varying the concentrations of HRP and H2O2. In this review, we outline the versatile properties of HRP-catalysed in situ-forming hydrogels, with a brief introduction to the crosslinking mechanisms involved. In addition, the recent biomedical applications of HRP-catalysed in situ-forming hydrogels for tissue regeneration are described.
Collapse
Affiliation(s)
- Jin Woo Bae
- Department of Molecular Science and Technology, Ajou University, Woncheon, Yeongtong, Suwon, Republic of Korea
| | - Jong Hoon Choi
- Department of Molecular Science and Technology, Ajou University, Woncheon, Yeongtong, Suwon, Republic of Korea
| | - Yunki Lee
- Department of Molecular Science and Technology, Ajou University, Woncheon, Yeongtong, Suwon, Republic of Korea
| | - Ki Dong Park
- Department of Molecular Science and Technology, Ajou University, Woncheon, Yeongtong, Suwon, Republic of Korea
| |
Collapse
|
18
|
Santo VE, Gomes ME, Mano JF, Reis RL. Controlled release strategies for bone, cartilage, and osteochondral engineering--Part I: recapitulation of native tissue healing and variables for the design of delivery systems. TISSUE ENGINEERING. PART B, REVIEWS 2013; 19:308-26. [PMID: 23268651 PMCID: PMC3690094 DOI: 10.1089/ten.teb.2012.0138] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 12/11/2012] [Indexed: 12/12/2022]
Abstract
The potential of growth factors to stimulate tissue healing through the enhancement of cell proliferation, migration, and differentiation is undeniable. However, critical parameters on the design of adequate carriers, such as uncontrolled spatiotemporal presence of bioactive factors, inadequate release profiles, and supraphysiological dosages of growth factors, have impaired the translation of these systems onto clinical practice. This review describes the healing cascades for bone, cartilage, and osteochondral interface, highlighting the role of specific growth factors for triggering the reactions leading to tissue regeneration. Critical criteria on the design of carriers for controlled release of bioactive factors are also reported, focusing on the need to provide a spatiotemporal control over the delivery and presentation of these molecules.
Collapse
Affiliation(s)
- Vítor E. Santo
- 3Bs Research Group—Biomaterials, Biodegradables, and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Manuela E. Gomes
- 3Bs Research Group—Biomaterials, Biodegradables, and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - João F. Mano
- 3Bs Research Group—Biomaterials, Biodegradables, and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L. Reis
- 3Bs Research Group—Biomaterials, Biodegradables, and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
19
|
Brochhausen C, Sánchez N, Halstenberg S, Zehbe R, Watzer B, Schmitt VH, Hofmann A, Meurer A, Unger RE, Kirkpatrick CJ. Phenotypic redifferentiation and cell cluster formation of cultured human articular chondrocytes in a three-dimensional oriented gelatin scaffold in the presence of PGE2- first results of a pilot study. J Biomed Mater Res A 2013; 101:2374-82. [DOI: 10.1002/jbm.a.34538] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 08/21/2012] [Accepted: 09/05/2012] [Indexed: 11/11/2022]
|
20
|
Andrés-Bergós J, Tardio L, Larranaga-Vera A, Gómez R, Herrero-Beaumont G, Largo R. The increase in O-linked N-acetylglucosamine protein modification stimulates chondrogenic differentiation both in vitro and in vivo. J Biol Chem 2012; 287:33615-28. [PMID: 22859309 PMCID: PMC3460460 DOI: 10.1074/jbc.m112.354241] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 07/31/2012] [Indexed: 11/06/2022] Open
Abstract
Insulin is an inducer of chondrocyte hypertrophy and growth plate chondrogenesis, although the specific molecular mechanisms behind these effects are mostly unknown. Our aim was to investigate whether insulin-induced chondrocyte hypertrophy occurs through a modification in the amount of O-linked N-acetylglucosamine (O-GlcNAc)-modified proteins and in the expression of the key enzymes of this pathway, O-GlcNAc transferase and O-GlcNAcase (OGA). We also studied if O-GlcNAc accumulation per se, induced by an OGA inhibitor, was able to induce pre-hypertrophic chondrocyte differentiation both in vitro and in vivo. Insulin-induced differentiation of ATDC5 pre-chondrocytes occurred alongside a gradual increase in the accumulation of O-GlcNac-modified proteins (O-GlcNAcylated proteins), as well as an increase in the expression of O-GlcNAc transferase and OGA. In the absence of insulin, O-GlcNAc accumulation induced by thiamet-G, a specific OGA inhibitor, was able to increase the gene expression of differentiation markers, as well as the activity of MMP-2 and -9. Thiamet-G also activated pERK, p-JNK, and p-p38 and the O-GlcNAcylation of Akt. Thiamet-G administration to C57/bl mice induced a significant expansion in the growth plate height and in the hypertrophic zone height. Therefore, our results show that O-GlcNAc glycosylation has chondromodulating activity.
Collapse
Affiliation(s)
- Jessica Andrés-Bergós
- From the Joint and Bone Research Unit, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid 28040, Spain
| | - Lidia Tardio
- From the Joint and Bone Research Unit, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid 28040, Spain
| | - Ane Larranaga-Vera
- From the Joint and Bone Research Unit, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid 28040, Spain
| | - Rodolfo Gómez
- From the Joint and Bone Research Unit, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid 28040, Spain
| | - Gabriel Herrero-Beaumont
- From the Joint and Bone Research Unit, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid 28040, Spain
| | - Raquel Largo
- From the Joint and Bone Research Unit, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid 28040, Spain
| |
Collapse
|
21
|
Wang Y, Huang YC, Gertzman AA, Xie L, Nizkorodov A, Hyzy SL, Truncale K, Guldberg RE, Schwartz Z, Boyan BD. Endogenous regeneration of critical-size chondral defects in immunocompromised rat xiphoid cartilage using decellularized human bone matrix scaffolds. Tissue Eng Part A 2012; 18:2332-42. [PMID: 22731693 DOI: 10.1089/ten.tea.2011.0688] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Clinical efforts to repair cartilage defects delivering cells or engineered cartilage implants into the lesions have met with limited success. This study used a critical-size chondral defect model in immunocompromised rat xiphoid cartilage to test whether endogenous chondrogenesis could be achieved using human bone matrix scaffolds to deliver human cartilage particles and/or a variant isoform of fibroblast growth factor-2 (FGF2-variant). Seventy-two male athymic RNU rats were enrolled in this study with eight rats per experimental group. Decellularized and demineralized human bone matrix scaffolds loaded with human articular cartilage particles or heat-inactivated cartilage particles were combined with different doses of the FGF2-variant. Scaffolds were implanted into 3-mm-diameter critical-size defects prepared using a biopsy punch through the center of the xiphoid. The samples were evaluated 28 days postsurgery using X-ray, equilibrium partitioning of ionic contrast microcomputed tomography, and safranin O-stained histological sagittal sections. Scaffolds containing cartilage particles plus the FGF2-variant induced dose-dependent increases in the formation of neocartilage (p<0.05), which was distributed homogeneously throughout the defects in comparison to scaffolds containing only the FGF2-variant. These effects were less pronounced when scaffolds with heat-inactivated cartilage particles were used. These results demonstrate that endogenous repair of chondral defects can be achieved in the absence of exogenous cells or bone marrow, suggesting that a similar approach may be successful for treating chondral lesions clinically.
Collapse
Affiliation(s)
- Yun Wang
- Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332-0363, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Santo VE, Gomes ME, Mano JF, Reis RL. Chitosan-chondroitin sulphate nanoparticles for controlled delivery of platelet lysates in bone regenerative medicine. J Tissue Eng Regen Med 2012; 6 Suppl 3:s47-59. [PMID: 22684916 DOI: 10.1002/term.1519] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 03/06/2012] [Indexed: 12/12/2022]
Abstract
In this study, a new formulation of nanoparticles (NPs) based on the electrostatic interaction between chitosan and chondroitin sulphate (CH-CS NPs) is proposed for the controlled release of proteins and growth factors (GFs), specifically platelet lysates (PLs). These nanoparticulate carriers are particularly promising for protein entrapment because the interactions between the polysaccharides and the entrapped proteins mimic the interactions between chondroitin sulphate and proteins in the native extracellular matrix (ECM). Spherical non-cytotoxic NPs were successfully produced, exhibiting high encapsulation efficiency for physiological levels of GFs and a controlled protein release profile for > 1 month. Moreover, it was also observed that these NPs can be uptaken by human adipose-derived stem cells (hASCs), depending on the concentration of NPs in the culture medium and incubation time. This shows the versatility of the developed NPs, which, besides acting as a protein delivery system, can also be used in the future as intracellular carriers for bioactive agents, such as nucleotides. When the PL-loaded NPs were used as a replacement of bovine serum for in vitro hASCs culture, the viability and proliferation of hASCs was not compromised. The release of PLs from CH-CS NPs also proved to be effective for the enhancement of in vitro osteogenic differentiation of hASCs, as shown by the increased levels of mineralization, suggesting not only the effective role of the delivery system but also the role of PLs as an osteogenic supplement for bone tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Vítor E Santo
- 3Bs Research Group - Biomaterials, Biodegradables and Biomimetics, Department of Polymer Engineering, University of Minho, Ave Park, Guimarães, Portugal
| | | | | | | |
Collapse
|
23
|
Freyria AM, Mallein-Gerin F. Chondrocytes or adult stem cells for cartilage repair: the indisputable role of growth factors. Injury 2012; 43:259-65. [PMID: 21696723 DOI: 10.1016/j.injury.2011.05.035] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 05/25/2011] [Indexed: 02/02/2023]
Abstract
Articular cartilage is easily injured but difficult to repair and cell therapies are proposed as tools to regenerate the defects in the tissue. Both differentiated chondrocytes and adult mesenchymal stem cells (MSCs) are regarded as cells potentially able to restore a functional cartilage. However, it is a complex process from the cell level to the tissue end product, during which growth factors play important roles from cell proliferation, extracellular matrix synthesis, maintenance of the phenotype to induction of MSCs towards chondrogenesis. Members of the TGF-β superfamily, are especially important in fulfilling these roles. Depending on the cell type chosen to restore cartilage, the effect of growth factors will vary. In this review, the roles of these factors in the maintenance of the chondrocyte phenotype are discussed and compared with those of factors involved in the repair of cartilage defects, using chondrocytes or adult mesenchymal stem cells.
Collapse
Affiliation(s)
- Anne-Marie Freyria
- Cartilage Biology and Engineering Group, IBCP, Université Lyon 1, Univ Lyon, CNRS FRE 3310, IFR128, France.
| | | |
Collapse
|
24
|
Hellingman CA, Koevoet W, van Osch GJVM. Can one generate stable hyaline cartilage from adult mesenchymal stem cells? A developmental approach. J Tissue Eng Regen Med 2011; 6:e1-e11. [DOI: 10.1002/term.502] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Revised: 05/27/2011] [Accepted: 07/13/2011] [Indexed: 01/30/2023]
Affiliation(s)
- Catharine A. Hellingman
- Department of Otorhinolaryngology, Head and Neck Surgery; Erasmus MC, University Medical Centre Rotterdam; PO Box 2040; 3000; CA; Rotterdam; The Netherlands
| | - Wendy Koevoet
- Department of Otorhinolaryngology, Head and Neck Surgery; Erasmus MC, University Medical Centre Rotterdam; PO Box 2040; 3000; CA; Rotterdam; The Netherlands
| | | |
Collapse
|
25
|
Challa TD, Rais Y, Ornan EM. Effect of adiponectin on ATDC5 proliferation, differentiation and signaling pathways. Mol Cell Endocrinol 2010; 323:282-91. [PMID: 20380870 DOI: 10.1016/j.mce.2010.03.025] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2010] [Revised: 03/28/2010] [Accepted: 03/30/2010] [Indexed: 12/18/2022]
Abstract
Adiponectin, an adipose-secreted adipocytokine, exhibits various metabolic functions but has no known effect on bone development through the growth plate and specifically, in chondrocytes. Using the mouse ATDC5 cell line, a widely used in vitro model of chondrogenesis, we demonstrated the expression of adiponectin and its receptors during chondrogenic differentiation. Adiponectin at 0.5mug/ml increased chondrocyte proliferation, proteoglycan synthesis and matrix mineralization, as reflected by upregulation of the expression of type II collagen, aggrecan, Runx2 and type X collagen, and of alkaline phosphatase activity. Quantitative RT-PCR and gelatin zymography showed a significant increase in the matrix metalloproteinase MMP9's expression and activity following adiponectin treatment. We therefore concluded that adiponectin can directly stimulate chondrocyte proliferation and differentiation. To evaluate the underlying mechanisms, we examined the effect of adiponectin on the expression of chondrogenic signaling molecules: Ihh, PTHrP, Ptc1, FGF18, BMP7, IGF1 and p21 were all upregulated while FGF9 was downregulated. This study reveals novel and direct activity of adiponectin in chondrocytes, suggesting its positive effects on bone development.
Collapse
Affiliation(s)
- T Delessa Challa
- Institute of Biochemistry and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Israel
| | | | | |
Collapse
|
26
|
Wachstumsfaktoren und Signalmoleküle zur Anwendung im „Tissue Engineering“. DER ORTHOPADE 2009; 38:1053-62. [DOI: 10.1007/s00132-009-1496-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
27
|
Wang J, Conboy I. Embryonic vs. adult myogenesis: challenging the 'regeneration recapitulates development' paradigm. J Mol Cell Biol 2009; 2:1-4. [PMID: 19797316 DOI: 10.1093/jmcb/mjp027] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
A popular theory in the stem cell field is that 'regeneration recapitulates development', or that adult stem cells function similarly to embryonic ones. In a recent Nature article, Lepper et al. questioned this approach, highlighting the differences in requirements for Pax7 during myogenesis for embryonic, juvenile and adult muscle.
Collapse
Affiliation(s)
- John Wang
- Department of Bioengineering, University of California, Berkeley, 174 Stanley Hall, Berkeley, CA 94720-1762, USA
| | | |
Collapse
|