251
|
Olvera D, Sathy BN, Kelly DJ. Spatial Presentation of Tissue-Specific Extracellular Matrix Components along Electrospun Scaffolds for Tissue Engineering the Bone-Ligament Interface. ACS Biomater Sci Eng 2020; 6:5145-5161. [PMID: 33455265 DOI: 10.1021/acsbiomaterials.0c00337] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The bone-ligament interface transitions from a highly organized type I collagen rich matrix to a nonmineralized fibrocartilage region and finally to a mineralized fibrocartilage region that interfaces with the bone. Therefore, engineering the bone-ligament interface requires a biomaterial substrate capable of maintaining or directing the spatially defined differentiation of multiple cell phenotypes. To date the appropriate combination of biophysical and biochemical factors that can be used to engineer such a biomaterial substrate remain unknown. Here we show that microfiber scaffolds functionalized with tissue-specific extracellular matrix (ECM) components can direct the differentiation of MSCs toward the phenotypes seen at the bone-ligament interface. Ligament-ECM (L-ECM) promoted the expression of the ligament-marker gene tenomodulin (TNMD) and higher levels of type I and III collagen expression compared to functionalization with commercially available type I collagen. Functionalization of microfiber scaffolds with cartilage-ECM (C-ECM) promoted chondrogenesis of MSCs, as evidenced by adoption of a round cell morphology and increased SRY-box 9 (SOX9) expression in the absence of exogenous growth factors. Next, we fabricated a multiphasic scaffold by controlling the spatial presentation of L-ECM and C-ECM along the length of a single electrospun microfiber construct, with the distal region of the C-ECM coated fibers additionally functionalized with an apatite layer (using simulated body fluid) to promote endochondral ossification. These ECM functionalized scaffolds promoted spatially defined differentiation of MSCs, with higher expression of TNMD observed in the region functionalized with L-ECM, and higher expression of type X collagen and osteopontin (markers of endochondral ossification) observed at the end of the scaffold functionalized with C-ECM and the apatite coating. Our results demonstrate the utility of tissue-specific ECM derived components as a cue for directing MSC differentiation when engineering complex multiphasic interfaces such as the bone-ligament enthesis.
Collapse
Affiliation(s)
- Dinorath Olvera
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.,Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
| | - Binulal N Sathy
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.,Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland.,Centre for Nanosciences & Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - Daniel J Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.,Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland.,Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
252
|
Neefjes M, van Caam APM, van der Kraan PM. Transcription Factors in Cartilage Homeostasis and Osteoarthritis. BIOLOGY 2020; 9:biology9090290. [PMID: 32937960 PMCID: PMC7563835 DOI: 10.3390/biology9090290] [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: 07/27/2020] [Revised: 09/07/2020] [Accepted: 09/07/2020] [Indexed: 12/13/2022]
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease, and it is characterized by articular cartilage loss. In part, OA is caused by aberrant anabolic and catabolic activities of the chondrocyte, the only cell type present in cartilage. These chondrocyte activities depend on the intra- and extracellular signals that the cell receives and integrates into gene expression. The key proteins for this integration are transcription factors. A large number of transcription factors exist, and a better understanding of the transcription factors activated by the various signaling pathways active during OA can help us to better understand the complex etiology of OA. In addition, establishing such a profile can help to stratify patients in different subtypes, which can be a very useful approach towards personalized therapy. In this review, we discuss crucial transcription factors for extracellular matrix metabolism, chondrocyte hypertrophy, chondrocyte senescence, and autophagy in chondrocytes. In addition, we discuss how insight into these factors can be used for treatment purposes.
Collapse
|
253
|
Li J, Sasaki N, Itaka K, Terpstra M, Levato R, Matsusaki M. Regulation of Chondrocyte Differentiation by Changing Intercellular Distances Using Type II Collagen Microfibers. ACS Biomater Sci Eng 2020; 6:5711-5719. [PMID: 33320579 DOI: 10.1021/acsbiomaterials.0c00427] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Osteoarthritis is a common degenerative disease that mainly occurs in older age groups, and the search for an effective cure remains a major global challenge. The technology of constructing 3D in vitro cartilage tissue with zonal differentiated structures for use as alternative implants for treating osteoarthritis has attracted researchers' attention. For this challenge, it is important for understanding the relationship between chondrocyte differentiation and the amount of extracellular matrix by modulating intercellular distance. This study investigates the interplay between chondrocyte differentiation and intercellular distance. Type II collagen microfibers (CMF II) were used as a distance regulator by varying their amounts. The results indicated that the secretion of cartilage-specific glycosaminoglycan after 2 weeks of differentiation from the chondrogenic cells, ATDC5, was decreased with an increased intercellular distance. Also, the shortest intercellular distance, being ATDC5 cells without CMF II, presented an upregulated gene expression profile of cartilage markers. The groups with CMF II-mediated intracellular distances, however, did not show the upregulation. The elastic modulus of the 3D samples increased depending on the amount of CMF II, relating to the differentiation preventing property of the CMF II. These findings suggest the promising potential of this approach for the modulation of chondrocyte differentiation.
Collapse
Affiliation(s)
- Jinyu Li
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
| | - Naoko Sasaki
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
| | - Keiji Itaka
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo 101-0062, Japan
| | - Margo Terpstra
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht 3584CX, The Netherlands
| | - Riccardo Levato
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht 3584CX, The Netherlands.,Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584CM, The Netherlands
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan.,Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
254
|
Salhotra A, Shah HN, Levi B, Longaker MT. Mechanisms of bone development and repair. Nat Rev Mol Cell Biol 2020; 21:696-711. [PMID: 32901139 DOI: 10.1038/s41580-020-00279-w] [Citation(s) in RCA: 620] [Impact Index Per Article: 124.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2020] [Indexed: 12/19/2022]
Abstract
Bone development occurs through a series of synchronous events that result in the formation of the body scaffold. The repair potential of bone and its surrounding microenvironment - including inflammatory, endothelial and Schwann cells - persists throughout adulthood, enabling restoration of tissue to its homeostatic functional state. The isolation of a single skeletal stem cell population through cell surface markers and the development of single-cell technologies are enabling precise elucidation of cellular activity and fate during bone repair by providing key insights into the mechanisms that maintain and regenerate bone during homeostasis and repair. Increased understanding of bone development, as well as normal and aberrant bone repair, has important therapeutic implications for the treatment of bone disease and ageing-related degeneration.
Collapse
Affiliation(s)
- Ankit Salhotra
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Harsh N Shah
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Benjamin Levi
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| | - Michael T Longaker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA. .,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
255
|
Anti-inflammatory capacity of Apremilast in human chondrocytes is dependent on SOX-9. Inflamm Res 2020; 69:1123-1132. [DOI: 10.1007/s00011-020-01392-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 07/28/2020] [Accepted: 08/08/2020] [Indexed: 12/12/2022] Open
|
256
|
Wang Z, Zhou W, Zheng G, Yang G. Inhibition of GPR17 with pranlukast protects against TNF-α-induced loss of type II collagen in ATDC5 cells. Int Immunopharmacol 2020; 88:106870. [PMID: 32805694 DOI: 10.1016/j.intimp.2020.106870] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 07/20/2020] [Accepted: 07/31/2020] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is a common joint disease affecting millions of elderly people worldwide. However, the mechanism of OA is complicated and remains poorly understood. Thus, a safe and effective therapeutic strategy has yet to be developed. G protein-coupled receptor 17 (GPR17) is an orphan receptor that is widely distributed in the central nervous system (CNS). GPR17 has become a target for the treatment of inflammation in brain diseases. In this study, we demonstrate that GPR17 is expressed in ATDC5 cells and is increased in response to TNF-α exposure. We also found that antagonism of GPR17 with pranlukast significantly inhibited oxidative stress by downregulating the intracellular level of reactive oxygen species (ROS) and increasing the activity of super oxide dismutase (SOD) against TNF-α. Interestingly, treatment with pranlukast prevented TNF-α-induced reduction of type II collagen. Additionally, knockdown of GPR17 with siRNA ameliorated TNF-α-induced loss of type II collagen, suggesting the importance of the role of GPR17 in mediating the impairment of type II collagen. Blockage of GPR17 with pranlukast suppressed the expression of matrix metalloproteinases 3 (MMP-3) and matrix metalloproteinases 13 (MMP-13), which contribute to the degradation of type II collagen. Pranlukast also prevented the activation of the JAK2/STAT1/IRF-1 signaling pathway, thereby suppressing the expression of pro-inflammatory cytokines and enzymes. Furthermore, pranlukast rescued TNF-α-induced reduced SOX-9 expression. Together, our data indicate that GPR17 might be a potential target for the treatment of OA.
Collapse
Affiliation(s)
- Zhangfu Wang
- Department of Spine Surgery, Taizhou Hospital of Wenzhou Medical University, Linhai, Taizhou 317000, China
| | - Weiwei Zhou
- Department of Spine Surgery, Taizhou Hospital of Wenzhou Medical University, Linhai, Taizhou 317000, China
| | - Guangbin Zheng
- Department of Spine Surgery, Taizhou Hospital of Wenzhou Medical University, Linhai, Taizhou 317000, China
| | - Guangyong Yang
- Department of Spine Surgery, Taizhou Hospital of Wenzhou Medical University, Linhai, Taizhou 317000, China.
| |
Collapse
|
257
|
An immortalized human adipose-derived stem cell line with highly enhanced chondrogenic properties. Biochem Biophys Res Commun 2020; 530:252-258. [PMID: 32828295 DOI: 10.1016/j.bbrc.2020.07.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/04/2020] [Indexed: 12/28/2022]
Abstract
Human adipose-derived stem cells (ASCs) are a commonly used cell type for cartilage tissue engineering. However, donor-to-donor variability, cell heterogeneity, inconsistent chondrogenic potential, and limited expansion potential can hinder the use of these cells for modeling chondrogenesis, in vitro screening of drugs and treatments for joint diseases, or translational applications for tissue engineered cartilage repair. The goal of this study was to create an immortalized ASC line that showed enhanced and consistent chondrogenic potential for applications in cartilage tissue engineering as well as to provide a platform for investigation of biological and mechanobiological pathways involved in cartilage homeostasis and disease. Starting with the ASC52telo cell line, a hTERT-immortalized ASC line, we used lentivirus to overexpress SOX9, a master regulator of chondrogenesis, and screened several clonal populations of SOX9 overexpressing cells to form a new stable cell line with high chondrogenic potential. One clonal line, named ASC52telo-SOX9, displayed increased GAG and type II collagen synthesis and was found to be responsive to both mechanical and inflammatory stimuli in a manner similar to native chondrocytes. The development of a clonal line such as ASC52telo-SOX9 has the potential to be a powerful tool for studying cartilage homeostasis and disease mechanisms in vitro, and potentially as a platform for in vitro drug screening for diseases that affect articular cartilage. Our findings provide an approach for the development of other immortalized cell lines with improved chondrogenic capabilities in ASCs or other adult stem cells.
Collapse
|
258
|
Nakamichi R, Kurimoto R, Tabata Y, Asahara H. Transcriptional, epigenetic and microRNA regulation of growth plate. Bone 2020; 137:115434. [PMID: 32422296 PMCID: PMC7387102 DOI: 10.1016/j.bone.2020.115434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/13/2020] [Accepted: 05/13/2020] [Indexed: 11/22/2022]
Abstract
Endochondral ossification is a critical event in bone formation, particularly in long shaft bones. Many cellular differentiation processes work in concert to facilitate the generation of cartilage primordium to formation of trabecular structures, all of which occur within the growth plate. Previous studies have revealed that the growth plate is tightly regulated by various transcription factors, epigenetic systems, and microRNAs. Hence, understanding these mechanisms that regulate the growth plate is crucial to furthering the current understanding on skeletal diseases, and in formulating effective treatment strategies. In this review, we focus on describing the function and mechanisms of the transcription factors, epigenetic systems, and microRNAs known to regulate the growth plate.
Collapse
Affiliation(s)
- Ryo Nakamichi
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, MBB-102, La Jolla, CA 92037, USA; Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Ryota Kurimoto
- Department of Systems Biomedicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yusuke Tabata
- Department of Orthopaedic Surgery, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Hirosi Asahara
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, MBB-102, La Jolla, CA 92037, USA; Department of Systems Biomedicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan.
| |
Collapse
|
259
|
Dash S, Trainor PA. The development, patterning and evolution of neural crest cell differentiation into cartilage and bone. Bone 2020; 137:115409. [PMID: 32417535 DOI: 10.1016/j.bone.2020.115409] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 05/04/2020] [Indexed: 12/12/2022]
Abstract
Neural crest cells are a vertebrate-specific migratory, multipotent cell population that give rise to a diverse array of cells and tissues during development. Cranial neural crest cells, in particular, generate cartilage, bone, tendons and connective tissue in the head and face as well as neurons, glia and melanocytes. In this review, we focus on the chondrogenic and osteogenic potential of cranial neural crest cells and discuss the roles of Sox9, Runx2 and Msx1/2 transcription factors and WNT, FGF and TGFβ signaling pathways in regulating neural crest cell differentiation into cartilage and bone. We also describe cranioskeletal defects and disorders arising from gain or loss-of-function of genes that are required for patterning and differentiation of cranial neural crest cells. Finally, we discuss the evolution of skeletogenic potential in neural crest cells and their function as a conduit for intraspecies and interspecies variation, and the evolution of craniofacial novelties.
Collapse
Affiliation(s)
- Soma Dash
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, MO, USA; Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
260
|
Vincent E, Villiard E, Sader F, Dhakal S, Kwok BH, Roy S. BMP signaling is essential for sustaining proximo-distal progression in regenerating axolotl limbs. Development 2020; 147:dev.170829. [PMID: 32665245 DOI: 10.1242/dev.170829] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 06/30/2020] [Indexed: 02/04/2023]
Abstract
Amputation of a salamander limb triggers a regeneration process that is perfect. A limited number of genes have been studied in this context and even fewer have been analyzed functionally. In this work, we use the BMP signaling inhibitor LDN193189 on Ambystoma mexicanum to explore the role of BMPs in regeneration. We find that BMP signaling is required for proper expression of various patterning genes and that its inhibition causes major defects in the regenerated limbs. Fgf8 is downregulated when BMP signaling is blocked, but ectopic injection of either human or axolotl protein did not rescue the defects. By administering LDN193189 treatments at different time points during regeneration, we show clearly that limb regeneration progresses in a proximal to distal fashion. This demonstrates that BMPs play a major role in patterning of regenerated limbs and that regeneration is a progressive process like development.
Collapse
Affiliation(s)
- Etienne Vincent
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, H3T 1J4, Canada
| | - Eric Villiard
- Department of Stomatology, Faculty of Dentistry, Université de Montréal, Montréal, Québec, H3T 1J4, Canada
| | - Fadi Sader
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, H3T 1J4, Canada
| | - Sabin Dhakal
- Institute for Research in Immunology and Cancer (IRIC), Département de médecine, Université de Montréal, Montréal, H3T 1J4, Canada
| | - Benjamin H Kwok
- Institute for Research in Immunology and Cancer (IRIC), Département de médecine, Université de Montréal, Montréal, H3T 1J4, Canada
| | - Stéphane Roy
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, H3T 1J4, Canada .,Department of Stomatology, Faculty of Dentistry, Université de Montréal, Montréal, Québec, H3T 1J4, Canada
| |
Collapse
|
261
|
Masaeli E, Nasr-Esfahani MH. An in vivo evaluation of induced chondrogenesis by decellularized extracellular matrix particles. J Biomed Mater Res A 2020; 109:627-636. [PMID: 32608181 DOI: 10.1002/jbm.a.37047] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 04/28/2020] [Accepted: 06/18/2020] [Indexed: 12/28/2022]
Abstract
Bioengineered scaffolds composed of synthetic materials and extracellular matrix (ECM) components can offer a tissue-specific microenvironment capable of regulating cells to regenerate the structure and function of the native cartilage. Here, given the potential preservation of biomechanical and biochemical cues found in the native cartilage, particulate decellularized ECM (DC-ECM) was utilized for immobilization on the surface of nanofibrous scaffolds. Afterward, the chondro-inductive potential and ectopic cartilage formation after subcutaneous implantation of bioengineered DC-ECM scaffolds were investigated in mice model. Eight weeks post-implantation, no growth of considerable inflammatory response and neovascularization was observed in histological images of bioengineered DC-ECM scaffolds. Pre-seeded bioengineered scaffolds with human adipose-derived stem cells exhibited high levels of chondro-induction capability, indicated with immunohistochemical and gene expression results. In both interval times, we also observed chondrogenesis and tissue formation after implanting unseeded bioengineered scaffolds, which denote that the presence of DC-ECM particles can even enhance attachment and migration of the host cells and induce chondrogenesis to them. To sum up, the incorporation of DC-ECM materials to tissue engineered constructs is a promising avenue to mimic the native tissue environment for regulation of cartilage regeneration in both in vivo and in vitro settings.
Collapse
Affiliation(s)
- Elahe Masaeli
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| |
Collapse
|
262
|
Zhu W, Yang X, Liu S, Wang M, Ye S, Luo H, Cui S. The involvement of cytokine-like 1 (Cytl1) in chondrogenesis and cartilage metabolism. Biochem Biophys Res Commun 2020; 529:608-614. [PMID: 32736681 DOI: 10.1016/j.bbrc.2020.06.069] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 06/15/2020] [Indexed: 11/26/2022]
Abstract
The Cytokine-like 1 (Cytl1) is first identified in bone marrow cells and preferentially expressed in cartilaginous tissue, and showed chondrogenic effects in mesenchymal cells, not essential for cartilage or bone development as in Cytl1 knock-out mice but associated with cartilage inflammatory and destruction. Here, we show the involvement of Cytl1 in chondrogenesis. Using specified chondrogenic embryonic skeleton and adult cartilage, the Cytl1 gene expression was investigated with associated chondrogenic factors by quantitative RT-PCR. The effect of Cytl1 protein (rCytl1) on cultured chondrocytes to regulate expression of key factors and phenotypic markers was studied. The results revealed that Cytl1 was highly expressed in chondrogenic process in embryos and adult cartilage. The rCytl1 increased the expression of Sox9 and Col2α1 with stabilized Col1α1 in cultured chondrocytes (redifferentiation). The Cytl1 was expressed and involved at all stages of cartilage development. Furthermore, Cytl1 expression shared similar patterns as other chondrogenic factors, implying interactions with other factors in chondrogenic process. Cytl1 is involved in cartilage development and matrix homeostasis, which defines the dedifferentiation phenotype of chondrocytes, essential to forming of functional cartilage in both physiologic remodeling and pathologic regeneration.
Collapse
Affiliation(s)
- Weicong Zhu
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Jinan University School of Medicine, Guangzhou, China
| | - Xiaohong Yang
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Jinan University School of Medicine, Guangzhou, China.
| | - Shaojie Liu
- Department of General Surgery, Guangzhou Red Cross Hospital, Jinan University School of Medicine, Guangzhou, China
| | - Min Wang
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Jinan University School of Medicine, Guangzhou, China
| | - Suihui Ye
- Department of General Surgery, Guangzhou Red Cross Hospital, Jinan University School of Medicine, Guangzhou, China
| | - Huixing Luo
- Department of General Surgery, Guangzhou Red Cross Hospital, Jinan University School of Medicine, Guangzhou, China
| | - Shuliang Cui
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Jinan University School of Medicine, Guangzhou, China; School of BioSciences, University of Melbourne, Victoria, Australia.
| |
Collapse
|
263
|
Veselis CA, Awan O, Thomas A, Ling S, Jonnalagadda P, Aneja A, Ali S. Bone Tumors Occurring in the Soft Tissues: A Review of the Clinical, Imaging, and Histopathologic Findings. Curr Probl Diagn Radiol 2020; 50:419-429. [PMID: 32665061 DOI: 10.1067/j.cpradiol.2020.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 06/07/2020] [Accepted: 06/22/2020] [Indexed: 12/16/2022]
Abstract
Although rare in everyday practice, malignancies that classically arise from bone or cartilage have been reported to arise de novo in various soft tissues in the body, resulting in a diagnostic challenge for the clinician, radiologist, and pathologist. Differential diagnoses of bone tumors often depend on anatomic location of the lesion. For example, the classic location of osteosarcoma is in the metaphysis of long bones about the knee. Histologically osteosarcoma is characterized by tumor cells that directly produce osteoid, bone, or cartilaginous matrix. In extraskeletal osteosarcoma, the clinical and radiologic picture is very different from a conventional osteosarcoma. They occur in older patients, present as a soft tissue mass often coincidentally following trauma and have a worse prognosis. The imaging characteristics are often nonspecific with mineralized elements in a well-defined soft tissue mass. The mineralized elements may or may not be visible. Magnetic Resonance sequences demonstrate a well circumscribed soft tissue mass with hemorrhagic and enhancing solid components. The pathologic features of extraskeletal osteosarcoma on a microscopic scale are identical to that of skeletal lesions. Likewise, conventional chondrosarcomas present in older patients with a growing, painful soft tissue prominence most commonly involving the long tubular bones. In extraskeletal chondrosarcoma however, the presentation is in somewhat younger patients with a painful soft tissue prominence typically in the head (meninges), neck, or upper leg. The pathologic features are most often that of a myxoid chondrosarcoma which is characterized by strands of small cells over a myxoid matrix. Imaging features include chondroid matrix, heterogenous contrast enhancement, and amorphous internal calcification on Computed Tomography. On Magnetic Resonance sequences the matrix has a low signal on all sequences, and variable inhomogeneity depending on grade of the lesion. Other extraskeletal bone tumors include Ewing's sarcoma and osteoid osteoma amongst other lesions. Although these malignancies may be rare clinical entities, they often exhibit characteristic clinical, imaging, and histopathological findings although differing in treatment and prognosis. Knowledge of these and other common mimicking lesions will help guide the clinician and radiologist to make an accurate diagnosis.
Collapse
Affiliation(s)
| | - Omer Awan
- University of Maryland School of Medicine. Baltimore, MD
| | | | | | | | | | - Sayed Ali
- Temple University Hospital. Philadelphia, PA
| |
Collapse
|
264
|
De Pieri A, Rana S, Korntner S, Zeugolis DI. Seaweed polysaccharides as macromolecular crowding agents. Int J Biol Macromol 2020; 164:434-446. [PMID: 32679331 DOI: 10.1016/j.ijbiomac.2020.07.087] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/30/2020] [Accepted: 07/09/2020] [Indexed: 02/07/2023]
Abstract
Development of mesenchymal stem cell-based tissue engineered implantable devices requires prolonged in vitro culture for the development of a three-dimensional implantable device, which leads to phenotypic drift, thus hindering the clinical translation and commercialisation of such approaches. Macromolecular crowding, a biophysical phenomenon based on the principles of excluded-volume effect, dramatically accelerates and increases extracellular matrix deposition during in vitro culture. However, the optimal macromolecular crowder is still elusive. Herein, we evaluated the biophysical properties of various concentrations of different seaweed in origin sulphated polysaccharides and their effect on human adipose derived stem cell cultures. Carrageenan, possibly due to its high sulphation degree, exhibited the highest negative charge values. No correlation was observed between the different concentrations of the crowders and charge, polydispersity index, hydrodynamic radius and fraction volume occupancy across all crowders. None of the crowders, but arabinogalactan, negatively affected cell viability. Carrageenan, fucoidan, galactofucan and ulvan increased extracellular matrix (especially collagen type I and collagen type V) deposition. Carrageenan induced the highest osteogenic effect and galactofucan and fucoidan demonstrated the highest chondrogenic effect. All crowders were relatively ineffective with respect to adipogenesis. Our data highlight the potential of sulphated seaweed polysaccharides for tissue engineering purposes.
Collapse
Affiliation(s)
- Andrea De Pieri
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Proxy Biomedical Ltd., Coilleach, Spiddal, Galway, Ireland
| | - Shubhasmin Rana
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Stefanie Korntner
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland.
| |
Collapse
|
265
|
Nakayama N, Pothiawala A, Lee JY, Matthias N, Umeda K, Ang BK, Huard J, Huang Y, Sun D. Human pluripotent stem cell-derived chondroprogenitors for cartilage tissue engineering. Cell Mol Life Sci 2020; 77:2543-2563. [PMID: 31915836 PMCID: PMC11104892 DOI: 10.1007/s00018-019-03445-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 12/24/2019] [Accepted: 12/27/2019] [Indexed: 02/06/2023]
Abstract
The cartilage of joints, such as meniscus and articular cartilage, is normally long lasting (i.e., permanent). However, once damaged, especially in large animals and humans, joint cartilage is not spontaneously repaired. Compensating the lack of repair activity by supplying cartilage-(re)forming cells, such as chondrocytes or mesenchymal stromal cells, or by transplanting a piece of normal cartilage, has been the basis of therapy for biological restoration of damaged joint cartilage. Unfortunately, current biological therapies face problems on a number of fronts. The joint cartilage is generated de novo from a specialized cell type, termed a 'joint progenitor' or 'interzone cell' during embryogenesis. Therefore, embryonic chondroprogenitors that mimic the property of joint progenitors might be the best type of cell for regenerating joint cartilage in the adult. Pluripotent stem cells (PSCs) are expected to differentiate in culture into any somatic cell type through processes that mimic embryogenesis, making human (h)PSCs a promising source of embryonic chondroprogenitors. The major research goals toward the clinical application of PSCs in joint cartilage regeneration are to (1) efficiently generate lineage-specific chondroprogenitors from hPSCs, (2) expand the chondroprogenitors to the number needed for therapy without loss of their chondrogenic activity, and (3) direct the in vivo or in vitro differentiation of the chondroprogenitors to articular or meniscal (i.e., permanent) chondrocytes rather than growth plate (i.e., transient) chondrocytes. This review is aimed at providing the current state of research toward meeting these goals. We also include our recent achievement of successful generation of "permanent-like" cartilage from long-term expandable, hPSC-derived ectomesenchymal chondroprogenitors.
Collapse
Affiliation(s)
- Naoki Nakayama
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA.
- Department of Orthopaedic Surgery, The University of Texas Health Science Center at Houston Medical School, Houston, TX, USA.
| | - Azim Pothiawala
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
| | - John Y Lee
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
- Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Nadine Matthias
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
| | - Katsutsugu Umeda
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
- Department of Pediatrics, Kyoto University School of Medicine, Kyoto, Japan
| | - Bryan K Ang
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
- Weil Cornell Medicine, New York, NY, USA
| | - Johnny Huard
- Department of Orthopaedic Surgery, The University of Texas Health Science Center at Houston Medical School, Houston, TX, USA
- Steadman Philippon Research Institute, Vail, CO, USA
| | - Yun Huang
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| | - Deqiang Sun
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| |
Collapse
|
266
|
Ledo AM, Vining KH, Alonso MJ, Garcia-Fuentes M, Mooney DJ. Extracellular matrix mechanics regulate transfection and SOX9-directed differentiation of mesenchymal stem cells. Acta Biomater 2020; 110:153-163. [PMID: 32417266 PMCID: PMC7291356 DOI: 10.1016/j.actbio.2020.04.027] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 12/21/2022]
Abstract
Gene delivery within hydrogel matrices can potentially direct mesenchymal stem cells (MSCs) towards a chondrogenic fate to promote regeneration of cartilage. Here, we investigated whether the mechanical properties of the hydrogel containing the gene delivery systems could enhance transfection and chondrogenic programming of primary human bone marrow-derived MSCs. We developed collagen-I-alginate interpenetrating polymer network hydrogels with tunable stiffness and adhesion properties. The hydrogels were activated with nanocomplexed SOX9 polynucleotides to direct chondrogenic differentiation of MSCs. MSCs transfected within the hydrogels showed higher expression of chondrogenic markers compared to MSCs transfected in 2D prior to encapsulation. The nanocomplex uptake and resulting expression of transfected SOX9 were jointly enhanced by increased stiffness and cell-adhesion ligand density in the hydrogels. Further, transfection of SOX9 effectively induced MSCs chondrogenesis and reduced markers of hypertrophy compared to control matrices. These findings highlight the importance of matrix stiffness and adhesion as design parameters in gene-activated matrices for regenerative medicine. STATEMENT OF SIGNIFICANCE: Gene-activated matrices (GAMs) are biodegradable polymer networks integrating gene therapies, and they are promising technologies for supporting tissue regeneration. Despite this interest, there is still limited information on how to rationally design these systems. Here, we provide a systematic study of the effect of matrix stiffness and cell adhesion ligands on gene transfer efficiency. We show that high stiffness and the presence of cell-binding sites promote transfection efficiency and that this result is related to more efficient internalization and trafficking of the gene therapies. GAMs with optimized mechanical properties can induce cartilage formation and result in tissues with better characteristics for articular cartilage tissue engineering as compared to previously described standard methods.
Collapse
Affiliation(s)
- Adriana M Ledo
- Department of Pharmacy and Pharmaceutical Technology, IDIS Research Institute, CIMUS Research Institute, University of Santiago de Compostela, Santiago de Compostela 15782, Spain.
| | - Kyle H Vining
- Wyss Institute for Biologically Inspired Engineering and John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.
| | - Maria J Alonso
- Department of Pharmacy and Pharmaceutical Technology, IDIS Research Institute, CIMUS Research Institute, University of Santiago de Compostela, Santiago de Compostela 15782, Spain.
| | - Marcos Garcia-Fuentes
- Department of Pharmacy and Pharmaceutical Technology, IDIS Research Institute, CIMUS Research Institute, University of Santiago de Compostela, Santiago de Compostela 15782, Spain.
| | - David J Mooney
- Wyss Institute for Biologically Inspired Engineering and John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
267
|
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
|
268
|
RNA-seq reveals downregulated osteochondral genes potentially related to tibia bacterial chondronecrosis with osteomyelitis in broilers. BMC Genet 2020; 21:58. [PMID: 32493207 PMCID: PMC7271470 DOI: 10.1186/s12863-020-00862-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 05/18/2020] [Indexed: 12/14/2022] Open
Abstract
Background Bacterial chondronecrosis with osteomyelitis (BCO) develops in the growth plate (GP) of the proximal femur and tibia and is initiated by damage to the less mineralized chondrocytes followed by colonization of opportunistic bacteria. This condition affects approximately 1% of all birds housed, being considered one of the major causes of lameness in fast growing broilers. Although several studies have been previously performed aiming to understand its pathogenesis, the molecular mechanisms involved with BCO remains to be elucidated. Therefore, this study aimed to generate a profile of global differential gene expression involved with BCO in the tibia of commercial broilers, through RNA sequencing analysis to identity genes and molecular pathways involved with BCO in chickens. Results Our data showed 192 differentially expressed (DE) genes: 63 upregulated and 129 downregulated in the GP of the tibia proximal epiphysis of BCO-affected broilers. Using all DE genes, six Biological Processes (BP) were associated with bone development (connective tissue development, cartilage development, skeletal system development, organ morphogenesis, system development and skeletal system morphogenesis). The analyses of the upregulated genes did not indicate any significant BP (FDR < 0.05). However, with the downregulated genes, the same BP were identified when using all DE genes in the analysis, with a total of 26 coding genes explaining BCO in the tibia: ACAN, ALDH1A2, CDH7, CHAD, CHADL, COL11A1, COMP, CSGALNACT1, CYR61, FRZB, GAL3ST1, HAPLN1, IHH, KIF26B, LECT1, LPPR1, PDE6B, RBP4A, SERINC5, SFRP1, SOX8, SOX9, TENM2, THBS1, UCHL1 and WFIKKN2. In addition, seven transcription factors were also associated to BCO: NFATC2, MAFB, HIF1A-ARNT, EWSR1-FLI1, NFIC, TCF3 and NF-KAPPAB. Conclusions Our data show that osteochondral downregulated genes are potential molecular causes of BCO in broilers, and the bacterial process seems to be, in fact, a secondary condition. Sixteen genes responsible for bone and cartilage formation were downregulated in BCO-affected broilers being strong candidate genes to trigger this disorder.
Collapse
|
269
|
Xu Y, Wang YQ, Wang AT, Yu CY, Luo Y, Liu RM, Zhao YJ, Xiao JH. Effect of CD44 on differentiation of human amniotic mesenchymal stem cells into chondrocytes via Smad and ERK signaling pathways. Mol Med Rep 2020; 21:2357-2366. [PMID: 32236637 PMCID: PMC7185282 DOI: 10.3892/mmr.2020.11044] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 02/28/2020] [Indexed: 01/22/2023] Open
Abstract
CD44 antigen (CD44) is a transmembrane protein found in cell adhesion molecules and is involved in the regulation of various physiological processes in cells. It was hypothesized that CD44 directly affected the chondrogenic differentiation of human amniotic mesenchymal stem cells (hAMSCs). In the present study, the expression of chondrocyte‑associated factors was detected in the absence and presence of the antibody blocker anti‑CD44 antibody during the chondrogenic differentiation of hAMSCs. Following inhibition of CD44 expression, the transcriptional levels of chondrocyte‑associated genes SRY‑box transcription factor 9, aggrecan and collagen type II α 1 chain, as well as the production of chondrocyte markers type II collagen and aggrecan were significantly decreased in hAMSCs. Further investigation indicated that there was no significant change in total ERK1/2 expression following inhibition of CD44 expression; however, phosphorylated (p)‑ERK1/2 expression was decreased. The expression of p‑Smad2/3 was also upregulated following CD44 inhibition. These data indicated that CD44 may affect the differentiation of hAMSCs into chondrocytes by regulating the Smad2/3 and ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Yan Xu
- Zunyi Municipal Key Laboratory of Medicinal Biotechnology, Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Yi-Qing Wang
- Zunyi Municipal Key Laboratory of Medicinal Biotechnology, Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Ai-Tong Wang
- Zunyi Municipal Key Laboratory of Medicinal Biotechnology, Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Chang-Yin Yu
- Center for Translational Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Yi Luo
- Zunyi Municipal Key Laboratory of Medicinal Biotechnology, Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
- Center for Translational Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Ru-Ming Liu
- Zunyi Municipal Key Laboratory of Medicinal Biotechnology, Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
- Center for Translational Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Yu-Jie Zhao
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Jian-Hui Xiao
- Zunyi Municipal Key Laboratory of Medicinal Biotechnology, Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
- Center for Translational Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
270
|
Semba JA, Mieloch AA, Rybka JD. Introduction to the state-of-the-art 3D bioprinting methods, design, and applications in orthopedics. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.bprint.2019.e00070] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
271
|
Bi R, Yin Q, Mei J, Chen K, Luo X, Fan Y, Zhu S. Identification of human temporomandibular joint fibrocartilage stem cells with distinct chondrogenic capacity. Osteoarthritis Cartilage 2020; 28:842-852. [PMID: 32147536 DOI: 10.1016/j.joca.2020.02.835] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 02/08/2020] [Accepted: 02/26/2020] [Indexed: 02/05/2023]
Abstract
OBJECTIVE This study was aimed to identify the residence of human fibrocartilage stem cells (hFCSCs), characterize their stem cell properties and investigate the functional mechanisms which regulate fibrocartilage stem cells (FCSCs) toward chondrogenic differentiation during cartilage homeostasis and repairing. METHODS Cytological characteristics of hFCSCs and human orofacial mesenchymal stem cells (hOFMSCs) were analyzed. Chondrogenic potential of hFCSCs was compared with hOFMSCs both in vitro and in vivo. Regulatory role of SOX9 during FCSCs chondrogenesis was studied by shRNA interference in vitro, and by GFP+ FCSCs treatment in rat condylar cartilage defect model. SOX9 expression was also examined in temporomandibular joint osteoarthritis (TMJOA) patients' cartilage surface. RESULTS hFCSCs exhibited typical mesenchymal stem cell characteristics, with significantly stronger chondrogenic capability compared to hOFMSCs. Moreover, hFCSCs showed remarkably increased expression of SOX9. During cartilage pellet culture, there was stronger SOX9 expression in hFCSCs than hOFMSCs. SOX9 shRNA interference downregulated chondrogenic capability of hFCSCs in vitro, as well as disrupting migration and chondrogenic differentiation of GFP+ FCSCs toward mature chondrocytes in rat condylar cartilage defect. Of note, SOX9 expression was also found suppressed in the condylar superficial zone of TMJOA patients. CONCLUSION We found the existence of FCSCs in human TMJ cartilage, and characterized their distinct stem cell features. SOX9 is essential for hFCSCs chondrogenic differentiation, and a comprehensive understanding of the regulatory role of SOX9 in hFCSCs would be important for exploring potential intervention strategy of condylar cartilage degradation during TMJ disorders.
Collapse
Affiliation(s)
- R Bi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Q Yin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - J Mei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - K Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - X Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Y Fan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - S Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
272
|
The RCAN1.4-calcineurin/NFAT signaling pathway is essential for hypoxic adaption of intervertebral discs. Exp Mol Med 2020; 52:865-875. [PMID: 32467610 PMCID: PMC7272636 DOI: 10.1038/s12276-020-0441-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 04/15/2020] [Indexed: 12/19/2022] Open
Abstract
Calcipressin-1, also known as regulator of calcineurin 1 (RCAN1), can specifically bind calcineurin at or near the calcineurin A catalytic domain and downregulate calcineurin activity. However, whether RCAN1 affects the hypoxic intervertebral disc (IVD) phenotype through the calcineurin/NFAT signaling pathway remains unclear. First, we confirmed the characteristics of the degenerative nucleus pulposus (NP) by H&E, safranin O/fast green and Alcian blue staining, and detected increased RCAN1 levels in the degenerative NP by immunohistochemistry. Then, we demonstrated that the protein level of RCAN1.4 was higher than that of RCAN1.1 and progressively elevated from the control group to the Pfirrmann grade V group. In vitro, both hypoxia (1% O2) and overexpression of HIF-1α reduced the protein level of RCAN1.4 in rat NP cells in a dose- and time-dependent manner. We further found that miRNA-124, through a nondegradative pathway (without the proteasome or lysosome), suppressed the expression of RCAN1.4. As expected, calcineurin in NP cells was activated and primarily promoted nuclear translocation of NFATc1 under hypoxia or RCAN1.4 siRNA transfection. Furthermore, SOX9, type II collagen and MMP13 were elevated under hypoxia, RCAN1.4 siRNA transfection or NFATc1 overexpression. Using chromatin immunoprecipitation (ChIP) and a luciferase reporter assay (with mutation), we clarified that NFATc1 increasingly bound the SOX9 promotor region (bp −367~−357). Interaction of HIF-1α and NFATc1 promoted MMP13 transcription. Finally, we found that FK506 reversed hypoxia-induced activation of the calcineurin/NFAT signaling pathway in NP cells and an ex vivo model. Together, these findings show that the RCAN1.4-calcineurin/NFAT signaling pathway has a vital role in the hypoxic phenotype of NP cells. RCAN1.4 might be a therapeutic target for degenerative disc diseases. Treatments targeting a protein that is overexpressed in damaged spinal cartilage could ease degenerative conditions associated with lower back pain. The intervertebral discs are complex cartilage tissues that absorb forces while allowing the motion of our spines. An immune-promoting enzyme called calcineurin is important in maintaining the supple, gel-like structure of the central part of each disc, the nucleus pulposus (NP). Fendong Zhao and Jian Chen at Zhejiang University School of Medicine Hangzhou, China and co-workers showed that RCAN1.4, a protein known to suppress calcineurin, is overexpressed in damaged human NPs. The team further revealed how a signaling pathway starting with RCAN1.4 suppresses key genes involved in forming the collagen fibers that hold the NP together. They therefore suggest that therapies targeting this protein could benefit patients suffering from disc degeneration diseases.
Collapse
|
273
|
Yang X, Liu TC, Liu S, Zhu W, Li H, Liang P, Ye S, Cui S. Promoted Viability and Differentiated Phenotype of Cultured Chondrocytes With Low Level Laser Irradiation Potentiate Efficacious Cells for Therapeutics. Front Bioeng Biotechnol 2020; 8:468. [PMID: 32548098 PMCID: PMC7272569 DOI: 10.3389/fbioe.2020.00468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 04/22/2020] [Indexed: 12/03/2022] Open
Abstract
Effective clinical treatments of cartilage lesions in affected joints require large numbers of viable chondrogenic cells generated through in vivo stimulation or ex vivo expansion of chondrocytes isolated from small biopsy specimens. Conventional passaging of chondrocytes in culture provides sufficient cells for treatments but these cells usually lose their differentiated phenotype. This leads to the formation of fibrocartilaginous tissue due to a malfunctioning repair process. Biostimulation of passaging chondrocytes with low level laser irradiation (LLLI) may theoretically produce more functional chondrocytes for cell-based repair of cartilage defects. Molecular and cellular analyses, cytochemistry, cell cultivation, and microscopy showed that LLLI treatments were found to (1) increase chondrocyte viability, (2) promote secretion of matrix proteins, (3) upregulate expression of chondrogenic genes, and (4) downregulate gene expression of cell destructive proteases and genes coding for mediators involved in the extrinsic apoptosis signaling pathway. Furthermore, LLLI attenuated induction of genes associated with cell death and matrix breakdown induced by IL-1β, some of which was seen at the protein level, with verification of effects on gene expression in the C28/I2 human chondrocyte line. LLLI treatments during culture generated larger numbers of viable chondrocytes compared to untreated cultures. Moreover, LLLI-treated chondrocytes in culture also rectified and simultaneously maintained their differentiated phenotype. Cultured chondrocytes treated with LLLI are a promising cell source for repairing cartilage lesions in vivo and restoration of articular function using tissue engineering strategies.
Collapse
Affiliation(s)
- Xiaohong Yang
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, School of Medicine, Jinan University, Guangzhou, China
| | - Timon Chengyi Liu
- Laboratory of Laser Sports Medicine, College of Physical Education and Sports Medicine, South China Normal University, Guangzhou, China
| | - Shaojie Liu
- Surgical Department, Guangzhou Red Cross Hospital, School of Medicine, Jinan University, Guangzhou, China
| | - Weicong Zhu
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, School of Medicine, Jinan University, Guangzhou, China
| | - Honglin Li
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, School of Medicine, Jinan University, Guangzhou, China
| | - Peihong Liang
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, School of Medicine, Jinan University, Guangzhou, China
| | - Suihui Ye
- Surgical Department, Guangzhou Red Cross Hospital, School of Medicine, Jinan University, Guangzhou, China
| | - Shuliang Cui
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, School of Medicine, Jinan University, Guangzhou, China.,School of BioSciences, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
274
|
Basu S, Mackowiak SD, Niskanen H, Knezevic D, Asimi V, Grosswendt S, Geertsema H, Ali S, Jerković I, Ewers H, Mundlos S, Meissner A, Ibrahim DM, Hnisz D. Unblending of Transcriptional Condensates in Human Repeat Expansion Disease. Cell 2020; 181:1062-1079.e30. [PMID: 32386547 PMCID: PMC7261253 DOI: 10.1016/j.cell.2020.04.018] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 01/16/2020] [Accepted: 04/13/2020] [Indexed: 11/27/2022]
Abstract
Expansions of amino acid repeats occur in >20 inherited human disorders, and many occur in intrinsically disordered regions (IDRs) of transcription factors (TFs). Such diseases are associated with protein aggregation, but the contribution of aggregates to pathology has been controversial. Here, we report that alanine repeat expansions in the HOXD13 TF, which cause hereditary synpolydactyly in humans, alter its phase separation capacity and its capacity to co-condense with transcriptional co-activators. HOXD13 repeat expansions perturb the composition of HOXD13-containing condensates in vitro and in vivo and alter the transcriptional program in a cell-specific manner in a mouse model of synpolydactyly. Disease-associated repeat expansions in other TFs (HOXA13, RUNX2, and TBP) were similarly found to alter their phase separation. These results suggest that unblending of transcriptional condensates may underlie human pathologies. We present a molecular classification of TF IDRs, which provides a framework to dissect TF function in diseases associated with transcriptional dysregulation.
Collapse
Affiliation(s)
- Shaon Basu
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Sebastian D Mackowiak
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Henri Niskanen
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Dora Knezevic
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Vahid Asimi
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Stefanie Grosswendt
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Hylkje Geertsema
- Institute for Chemistry and Biochemistry, Free University Berlin, 14195 Berlin, Germany
| | - Salaheddine Ali
- RG Development and Disease, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany; Institute for Medical and Human Genetics, Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; Charité-Universitätsmedizin Berlin, BCRT-Berlin Institute of Health Center for Regenerative Therapies, 10178 Berlin, Germany
| | - Ivana Jerković
- RG Development and Disease, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Helge Ewers
- Institute for Chemistry and Biochemistry, Free University Berlin, 14195 Berlin, Germany
| | - Stefan Mundlos
- RG Development and Disease, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany; Institute for Medical and Human Genetics, Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; Charité-Universitätsmedizin Berlin, BCRT-Berlin Institute of Health Center for Regenerative Therapies, 10178 Berlin, Germany
| | - Alexander Meissner
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Daniel M Ibrahim
- RG Development and Disease, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany; Institute for Medical and Human Genetics, Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; Charité-Universitätsmedizin Berlin, BCRT-Berlin Institute of Health Center for Regenerative Therapies, 10178 Berlin, Germany
| | - Denes Hnisz
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany.
| |
Collapse
|
275
|
Li L, Wei X, Wang D, Lv Z, Geng X, Li P, Lu J, Wang K, Wang X, Sun J, Cao X, Wei L. Positive Effects of a Young Systemic Environment and High Growth Differentiation Factor 11 Levels on Chondrocyte Proliferation and Cartilage Matrix Synthesis in Old Mice. Arthritis Rheumatol 2020; 72:1123-1133. [PMID: 32067417 DOI: 10.1002/art.41230] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 02/06/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To investigate the effects of a young systemic environment and growth differentiation factor 11 (GDF-11) on aging cartilage. METHODS A heterochronic parabiosis model (2-month-old mouse and 12-month-old mouse [Y/O]), an isochronic parabiosis model (12-month-old mouse and 12-month-old mouse [O/O]), and 12-month-old mice alone (O) were evaluated. Knee joints and chondrocytes from old mice were examined by radiography, histology, cell proliferation assays, immunohistochemistry, Western blotting, and quantitative reverse transcriptase-polymerase chain reaction 16 weeks after parabiosis surgery. GDF-11 was injected into 12-month-old mouse joints daily for 16 weeks. Cartilage degeneration, cell proliferation, and osteoarthritis-related gene expression were evaluated. RESULTS Osteoarthritis Research Society International scores in old mice were significantly lower in the Y/O group than in the O/O and O groups (both P < 0.05). The percentage of 5-ethynyl-2'-deoxyuridine-positive chondrocytes in old mice was significantly higher in the Y/O group than in the other groups (P < 0.05). Type II collagen (CII) and SOX9 messenger RNA levels differed in cartilage from old mice in the Y/O group compared to the O/O and O groups (both P < 0.05). RUNX-2, CX, and matrix metalloproteinase 13 levels were significantly lower in cartilage from old mice in the Y/O group compared to the O/O and O groups (both P < 0.05). Similar results were obtained for protein expression levels and after GDF-11 treatment in vitro and in vivo. Phosphorylated Smad2/3 (pSmad2/3) levels were higher in the recombinant GDF-11-treated group than in the control group. CONCLUSION A young systemic environment promotes chondrocyte proliferation and cartilage matrix synthesis in old mice. GDF-11, a "young factor," contributes to these effects through the up-regulation of pSmad2/3.
Collapse
Affiliation(s)
- Lu Li
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaochun Wei
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Dongming Wang
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhi Lv
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiang Geng
- Shanxi Health Vocational College, Jinzhong, China
| | - Pengcui Li
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jiangong Lu
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Kaihang Wang
- Subsidiary High School of Taiyuan Normal University, Taiyuan, China
| | - Xiaohu Wang
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jian Sun
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaoming Cao
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Lei Wei
- Warren Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
276
|
Hirano T, Saito D, Yamada H, Ishisaki A, Kamo M. TGF-β1 induces N-cadherin expression by upregulating Sox9 expression and promoting its nuclear translocation in human oral squamous cell carcinoma cells. Oncol Lett 2020; 20:474-482. [PMID: 32565972 PMCID: PMC7285821 DOI: 10.3892/ol.2020.11582] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 01/29/2020] [Indexed: 02/06/2023] Open
Abstract
Squamous cell carcinoma (SCC) is the most frequent cancer that develops in the oral cavity. Epithelial-mesenchymal transition (EMT) is known to play an important role in the process of metastasis of SCC cells. In our previous study, we demonstrated that TGF-β1 induced EMT in the human oral SCC (hOSCC) cell line HSC-4. We also found that Slug plays an important role in suppressing E-cadherin expression and promotion of the migratory activity of HSC-4 cells. However, we also demonstrated that Slug does not participate in upregulation of N-cadherin expression, suggesting that EMT-related transcription factors other than Slug also play an important role in the process. In the present study, we aimed to elucidate how the transcription factor Sox9 affects the TGF-β1-induced upregulation of N-cadherin expression in HSC-4 cells. We found that TGF-β1 upregulated Sox9 expression in HSC-4 cells. In addition, Sox9 siRNA significantly abrogated the TGF-β1-induced upregulation of N-cadherin expression and inhibited the TGF-β1-promoted migratory activity in HSC-4 cells. We also demonstrated that TGF-β1 upregulated the phosphorylation status of Sox9 and then promoted nuclear translocation of Sox9 from the cytoplasm, possibly resulting in an increase in N-cadherin expression. The cyclic AMP-dependent protein kinase A inhibitor H-89, which is known to suppress phosphorylation of Sox9, significantly abrogated the TGF-β1-induced upregulation of N-cadherin expression. These results suggested that TGF-β1 induced N-cadherin expression by upregulating Sox9 expression and promoting its nuclear translocation, which results in EMT progression in hOSCC cells.
Collapse
Affiliation(s)
- Taifu Hirano
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba-cho, Iwate 028-3694, Japan.,Division of Oral and Maxillofacial Surgery, Department of Reconstructive Oral and Maxillofacial Surgery, Iwate Medical University School of Dentistry, Morioka, Iwate 020-8505, Japan
| | - Daishi Saito
- Division of Oral and Maxillofacial Surgery, Department of Reconstructive Oral and Maxillofacial Surgery, Iwate Medical University School of Dentistry, Morioka, Iwate 020-8505, Japan
| | - Hiroyuki Yamada
- Division of Oral and Maxillofacial Surgery, Department of Reconstructive Oral and Maxillofacial Surgery, Iwate Medical University School of Dentistry, Morioka, Iwate 020-8505, Japan
| | - Akira Ishisaki
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba-cho, Iwate 028-3694, Japan
| | - Masaharu Kamo
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba-cho, Iwate 028-3694, Japan
| |
Collapse
|
277
|
Song H, Park KH. Regulation and function of SOX9 during cartilage development and regeneration. Semin Cancer Biol 2020; 67:12-23. [PMID: 32380234 DOI: 10.1016/j.semcancer.2020.04.008] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 09/23/2019] [Accepted: 04/26/2020] [Indexed: 12/21/2022]
Abstract
Chondrogenesis is a highly coordinated event in embryo development, adult homeostasis, and repair of the vertebrate cartilage. Fate decisions and differentiation of chondrocytes accompany differential expression of genes critical for each step of chondrogenesis. SOX9 is a master transcription factor that participates in sequential events in chondrogenesis by regulating a series of downstream factors in a stage-specific manner. SOX9 either works alone or in combination with downstream SOX transcription factors, SOX5 and SOX6 as chondrogenic SOX Trio. SOX9 is reduced in the articular cartilage of patients with osteoarthritis while highly maintained during tumorigenesis of cartilage and bone. Gene therapy using viral and non-viral vectors accompanied by tissue engineering (scaffolds) is a promising tool to regenerate impaired cartilage. Delivery of SOX9 or chondrogenic SOX Trio into cells produces efficient therapeutic effects on chondrogenesis and this event is facilitated by scaffolds. Non-viral vector-guided delivery systems encapsulated or loaded in mechanically stable solid scaffolds are useful for the regeneration of articular cartilage. Here we review major milestones and most recent studies focusing on regulation and function of chondrogenic SOX Trio, during chondrogenesis and cartilage regeneration, and on the development of advanced technologies in gene delivery with tissue engineering to improve efficiency of cartilage repair process.
Collapse
Affiliation(s)
- Haengseok Song
- Department of Biomedical Science, CHA University, Seongnam, Republic of Korea
| | - Keun-Hong Park
- Department of Biomedical Science, CHA University, Seongnam, Republic of Korea.
| |
Collapse
|
278
|
Meng W, Rey-Rico A, Claudel M, Schmitt G, Speicher-Mentges S, Pons F, Lebeau L, Venkatesan JK, Cucchiarini M. rAAV-Mediated Overexpression of SOX9 and TGF-β via Carbon Dot-Guided Vector Delivery Enhances the Biological Activities in Human Bone Marrow-Derived Mesenchymal Stromal Cells. NANOMATERIALS 2020; 10:nano10050855. [PMID: 32354138 PMCID: PMC7712756 DOI: 10.3390/nano10050855] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 12/21/2022]
Abstract
Scaffold-assisted gene therapy is a highly promising tool to treat articular cartilage lesions upon direct delivery of chondrogenic candidate sequences. The goal of this study was to examine the feasibility and benefits of providing highly chondroreparative agents, the cartilage-specific sex-determining region Y-type high-mobility group 9 (SOX9) transcription factor or the transforming growth factor beta (TGF-β), to human bone marrow-derived mesenchymal stromal cells (hMSCs) via clinically adapted, independent recombinant adeno-associated virus (rAAV) vectors formulated with carbon dots (CDs), a novel class of carbon-dominated nanomaterials. Effective complexation and release of a reporter rAAV-lacZ vector was achieved using four different CDs elaborated from 1-citric acid and pentaethylenehexamine (CD-1); 2-citric acid, poly(ethylene glycol) monomethyl ether (MW 550 Da), and N,N-dimethylethylenediamine (CD-2); 3-citric acid, branched poly(ethylenimine) (MW 600 Da), and poly(ethylene glycol) monomethyl ether (MW 2 kDa) (CD-3); and 4-citric acid and branched poly(ethylenimine) (MW 600 Da) (CD-4), allowing for the genetic modification of hMSCs. Among the nanoparticles, CD-2 showed an optimal ability for rAAV delivery (up to 2.2-fold increase in lacZ expression relative to free vector treatment with 100% cell viability for at least 10 days, the longest time point examined). Administration of therapeutic (SOX9, TGF-β) rAAV vectors in hMSCs via CD-2 led to the effective overexpression of each independent transgene, promoting enhanced cell proliferation (TGF-β) and cartilage matrix deposition (glycosaminoglycans, type-II collagen) for at least 21 days relative to control treatments (CD-2 lacking rAAV or associated to rAAV-lacZ), while advantageously restricting undesirable type-I and -X collagen deposition. These results reveal the potential of CD-guided rAAV gene administration in hMSCs as safe, non-invasive systems for translational strategies to enhance cartilage repair.
Collapse
Affiliation(s)
- Weikun Meng
- Center of Experimental Orthopaedics, Saarland University Medical Center, D-66421 Homburg, Germany
| | - Ana Rey-Rico
- Cell Therapy and Regenerative Medicine Unit, Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña, ES-15071 A Coruña, Spain
| | - Mickaël Claudel
- Laboratoire de Conception et Application de Molécules Bioactives, Faculty of Pharmacy, UMR 7199 CNRS—University of Strasbourg, F-67401 Illkirch, France
| | - Gertrud Schmitt
- Center of Experimental Orthopaedics, Saarland University Medical Center, D-66421 Homburg, Germany
| | - Susanne Speicher-Mentges
- Center of Experimental Orthopaedics, Saarland University Medical Center, D-66421 Homburg, Germany
| | - Françoise Pons
- Laboratoire de Conception et Application de Molécules Bioactives, Faculty of Pharmacy, UMR 7199 CNRS—University of Strasbourg, F-67401 Illkirch, France
| | - Luc Lebeau
- Laboratoire de Conception et Application de Molécules Bioactives, Faculty of Pharmacy, UMR 7199 CNRS—University of Strasbourg, F-67401 Illkirch, France
| | - Jagadeesh K. Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, D-66421 Homburg, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, D-66421 Homburg, Germany
- Correspondence: ; Tel.: +49-6841-1624-987; Fax: +49-6841-1624-988
| |
Collapse
|
279
|
Mahendra CK, Tan LTH, Lee WL, Yap WH, Pusparajah P, Low LE, Tang SY, Chan KG, Lee LH, Goh BH. Angelicin-A Furocoumarin Compound With Vast Biological Potential. Front Pharmacol 2020; 11:366. [PMID: 32372949 PMCID: PMC7176996 DOI: 10.3389/fphar.2020.00366] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 03/10/2020] [Indexed: 12/14/2022] Open
Abstract
Angelicin, a member of the furocoumarin group, is related to psoralen which is well known for its effectiveness in phototherapy. The furocoumarins as a group have been studied since the 1950s but only recently has angelicin begun to come into its own as the subject of several biological studies. Angelicin has demonstrated anti-cancer properties against multiple cell lines, exerting effects via both the intrinsic and extrinsic apoptotic pathways, and also demonstrated an ability to inhibit tubulin polymerization to a higher degree than psoralen. Besides that, angelicin too demonstrated anti-inflammatory activity in inflammatory-related respiratory and neurodegenerative ailments via the activation of NF-κB pathway. Angelicin also showed pro-osteogenesis and pro-chondrogenic effects on osteoblasts and pre-chondrocytes respectively. The elevated expression of pro-osteogenic and chondrogenic markers and activation of TGF-β/BMP, Wnt/β-catenin pathway confirms the positive effect of angelicin bone remodeling. Angelicin also increased the expression of estrogen receptor alpha (ERα) in osteogenesis. Other bioactivities, such as anti-viral and erythroid differentiating properties of angelicin, were also reported by several researchers with the latter even displaying an even greater aptitude as compared to the commonly prescribed drug, hydroxyurea, which is currently on the market. Apart from that, recently, a new application for angelicin against periodontitis had been studied, where reduction of bone loss was indirectly caused by its anti-microbial properties. All in all, angelicin appears to be a promising compound for further studies especially on its mechanism and application in therapies for a multitude of common and debilitating ailments such as sickle cell anaemia, osteoporosis, cancer, and neurodegeneration. Future research on the drug delivery of angelicin in cancer, inflammation and erythroid differentiation models would aid in improving the bioproperties of angelicin and efficacy of delivery to the targeted site. More in-depth studies of angelicin on bone remodeling, the pro-osteogenic effect of angelicin in various bone disease models and the anti-viral implications of angelicin in periodontitis should be researched. Finally, studies on the binding of angelicin toward regulatory genes, transcription factors, and receptors can be done through experimental research supplemented with molecular docking and molecular dynamics simulation.
Collapse
Affiliation(s)
- Camille Keisha Mahendra
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia
- Novel Bacteria and Drug Discovery Research Group, Microbiome and Bioresource Research Strength Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Subang Jaya, Malaysia
| | - Loh Teng Hern Tan
- Novel Bacteria and Drug Discovery Research Group, Microbiome and Bioresource Research Strength Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Subang Jaya, Malaysia
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Wai Leng Lee
- School of Science, Monash University Malaysia, Subang Jaya, Malaysia
| | - Wei Hsum Yap
- School of Biosciences, Taylor's University, Subang Jaya, Malaysia
| | - Priyia Pusparajah
- Medical Health and Translational Research Group, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
| | - Liang Ee Low
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China
| | - Siah Ying Tang
- Chemical Engineering Discipline, School of Engineering, Monash University Malaysia, Subang Jaya, Malaysia
- Advanced Engineering Platform, Monash University Malaysia, Subang Jaya, Malaysia
| | - Kok Gan Chan
- International Genome Centre, Jiangsu University, Zhenjiang, China
- Division of Genetics and Molecular Biology, Faculty of Science, Institute of Biological Sciences, University of Malaya, Kuala Lumpur, Malaysia
| | - Learn Han Lee
- Novel Bacteria and Drug Discovery Research Group, Microbiome and Bioresource Research Strength Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Subang Jaya, Malaysia
| | - Bey Hing Goh
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Health and Well-Being Cluster, Global Asia in the 21st Century (GA21) Platform, Monash University Malaysia, Subang Jaya, Malaysia
| |
Collapse
|
280
|
Matsuura VKSK, Yoshida CA, Komori H, Sakane C, Yamana K, Jiang Q, Komori T. Expression of a Constitutively Active Form of Hck in Chondrocytes Activates Wnt and Hedgehog Signaling Pathways, and Induces Chondrocyte Proliferation in Mice. Int J Mol Sci 2020; 21:E2682. [PMID: 32290615 PMCID: PMC7215647 DOI: 10.3390/ijms21082682] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 03/30/2020] [Accepted: 04/09/2020] [Indexed: 12/30/2022] Open
Abstract
Runx2 is required for chondrocyte proliferation and maturation. In the search of Runx2 target genes in chondrocytes, we found that Runx2 up-regulated the expression of hematopoietic cell kinase (Hck), which is a member of the Src tyrosine kinase family, in chondrocytes, that Hck expression was high in cartilaginous limb skeletons of wild-type mice but low in those of Runx2-/- mice, and that Runx2 bound the promoter region of Hck. To investigate the functions of Hck in chondrocytes, transgenic mice expressing a constitutively active form of Hck (HckCA) were generated using the Col2a1 promoter/enhancer. The hind limb skeletons were fused, the tibia became a large, round mass, and the growth plate was markedly disorganized. Chondrocyte maturation was delayed until E16.5 but accelerated thereafter. BrdU-labeled, but not terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL)-positive, chondrocytes were increased. Furthermore, Hck knock-down reduced the proliferation of primary chondrocytes. In microarray and real-time RT-PCR analyses using hind limb RNA from HckCA transgenic mice, the expression of Wnt (Wnt10b, Tcf7, Lef1, Dkk1) and hedgehog (Ihh, Ptch1, and Gli1) signaling pathway genes was upregulated. These findings indicated that Hck, whose expression is regulated by Runx2, is highly expressed in chondrocytes, and that HckCA activates Wnt and hedgehog signaling pathways, and promotes chondrocyte proliferation without increasing apoptosis.
Collapse
Affiliation(s)
- Viviane K. S. Kawata Matsuura
- Basic and Translational Research Center for Hard Tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Carolina Andrea Yoshida
- Basic and Translational Research Center for Hard Tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Hisato Komori
- Basic and Translational Research Center for Hard Tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Chiharu Sakane
- Division of Comparative Medicine, Life Science Support Center, Nagasaki University, Nagasaki 852-8523, Japan
| | - Kei Yamana
- Teijin Institute for Bio-Medical Research, TEIJIN LIMITED, Tokyo 100-8585, Japan
| | - Qing Jiang
- Basic and Translational Research Center for Hard Tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Toshihisa Komori
- Basic and Translational Research Center for Hard Tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| |
Collapse
|
281
|
Rice SJ, Beier F, Young DA, Loughlin J. Interplay between genetics and epigenetics in osteoarthritis. Nat Rev Rheumatol 2020; 16:268-281. [PMID: 32273577 DOI: 10.1038/s41584-020-0407-3] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2020] [Indexed: 12/15/2022]
Abstract
Research into the molecular genetics of osteoarthritis (OA) has been substantially bolstered in the past few years by the implementation of powerful genome-wide scans that have revealed a large number of novel risk loci associated with the disease. This refreshing wave of discovery has occurred concurrently with epigenetic studies of joint tissues that have examined DNA methylation, histone modifications and regulatory RNAs. These epigenetic analyses have involved investigations of joint development, homeostasis and disease and have used both human samples and animal models. What has become apparent from a comparison of these two complementary approaches is that many OA genetic risk signals interact with, map to or correlate with epigenetic mediators. This discovery implies that epigenetic mechanisms, and their effect on gene expression, are a major conduit through which OA genetic risk polymorphisms exert their functional effects. This observation is particularly exciting as it provides mechanistic insight into OA susceptibility. Furthermore, this knowledge reveals avenues for attenuating the negative effect of risk-conferring alleles by exposing the epigenome as an exploitable target for therapeutic intervention in OA.
Collapse
Affiliation(s)
- Sarah J Rice
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Frank Beier
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada.,Western Bone and Joint Institute, The University of Western Ontario, London, ON, Canada
| | - David A Young
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - John Loughlin
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
282
|
Wang H, Chen H, Chernick M, Li D, Ying GG, Yang J, Zheng N, Xie L, Hinton DE, Dong W. Selenomethionine exposure affects chondrogenic differentiation and bone formation in Japanese medaka (Oryzias latipes). JOURNAL OF HAZARDOUS MATERIALS 2020; 387:121720. [PMID: 31812480 DOI: 10.1016/j.jhazmat.2019.121720] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/07/2019] [Accepted: 11/18/2019] [Indexed: 06/10/2023]
Abstract
Excess selenium entering the aquatic environment from anthropogenic activities has been associated with developmental abnormalities in fish including skeletal deformities of the head and spine. However, mechanisms of this developmental toxicity have not been well-characterized. In this study, Japanese medaka (Oryzias latipes) embryos were exposed to seleno-l-methionine (Se-Met) in a range of concentrations. Gene expression was evaluated for sex-determining region Y (SRY)-related box (Sox9a and Sox9b), runt-related transcription factor 2 (Runx2), and melatonin receptor (Mtr). Alterations in the length of Meckel's cartilage, tail curvature, and decreased calcification were observed in skeletal stains at 10- and 22-days post-fertilization (dpf). Embryonic exposure of Osterix-mCherry transgenic medaka resulted in fewer teeth. Sox9a and Sox9b were up-regulated, while Runx2 and Mtr were down-regulated by Se-Met prior to hatch. Whole mount in situ hybridization (WISH) localized gene expression to areas observed to be affected in vivo. In addition, Se-Met exposures of a Mtr morpholino (Mtr-MO) as well as Luzindole exposed embryos developed similar skeletal malformations, supporting involvement of Mtr. These findings demonstrate that Se-Met modulates expression of key genes involved in chondrogenic differentiation and bone formation during development.
Collapse
Affiliation(s)
- Huan Wang
- College of Animal Science and Technology, Inner Mongolia University for Nationalities/Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, Tongliao, 028000, China
| | - Hongxing Chen
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou, 510006, China; School of Environment, South China Normal University, University Town, Guangzhou, 510006, China
| | - Melissa Chernick
- Nicholas School of the Environment, Duke University, Durham, NC, 27708, USA
| | - Dan Li
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou, 510006, China; School of Environment, South China Normal University, University Town, Guangzhou, 510006, China
| | - Guang-Guo Ying
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou, 510006, China; School of Environment, South China Normal University, University Town, Guangzhou, 510006, China
| | - Jingfeng Yang
- College of Animal Science and Technology, Inner Mongolia University for Nationalities/Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, Tongliao, 028000, China
| | - Na Zheng
- Nicholas School of the Environment, Duke University, Durham, NC, 27708, USA; Key Laboratory of Wetland Ecology and Environment, Northeast Institute of Geography and Agricultural Ecology, Chinese Academy of Sciences, Changchun, Jilin, 130012, China
| | - Lingtian Xie
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou, 510006, China; School of Environment, South China Normal University, University Town, Guangzhou, 510006, China.
| | - David E Hinton
- Nicholas School of the Environment, Duke University, Durham, NC, 27708, USA.
| | - Wu Dong
- College of Animal Science and Technology, Inner Mongolia University for Nationalities/Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, Tongliao, 028000, China; Nicholas School of the Environment, Duke University, Durham, NC, 27708, USA.
| |
Collapse
|
283
|
Liu ST, Chou MY, Wu LC, Horng JL, Lin LY. Transient receptor potential vanilloid 4 modulates ion balance through the isotocin pathway in zebrafish (Danio rerio). Am J Physiol Regul Integr Comp Physiol 2020; 318:R751-R759. [DOI: 10.1152/ajpregu.00307.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Isotocin controls ion regulation through modulating the functions of ionocytes (also called mitochondria-rich cells or chloride cells). However, little is known about the upstream molecule of the isotocin system. Herein, we identify transient receptor potential vanilloid 4 (TRPV4), which regulates the mRNA and protein expressions of isotocin and affects ion regulation through the isotocin pathway. Double immunohistochemical results showed that TRPV4 is expressed in isotocinergic neurons in the hypothalamus of the adult zebrafish brain. To further elucidate the roles of TRPV4, we manipulated TRPV4 protein expression and evaluated its ionoregulatory functions in zebrafish embryos. TRPV4 gene knockdown with morpholino oligonucleotides decreased ionic contents (Na+, Cl−, and Ca2+) of whole larvae and the H+-secreting function of larval skin of zebrafish. mRNA expressions of ionocyte-related transporters, including H+-ATPase, the epithelial Ca2+ channel, and the Na+-Cl− cotransporter, were also suppressed in trpv4 morphants. Numbers of ionocytes (H+-ATPase-rich cells and Na+-K+-ATPase-rich cells) and epidermal stem cells in zebrafish larval skin also decreased after trpv4 knockdown. Our results showed that TRPV4 modulates ion balance through the isotocin pathway.
Collapse
Affiliation(s)
- Sian-Tai Liu
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Ming-Yi Chou
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Liang-Chun Wu
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Jiun-Lin Horng
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Li-Yih Lin
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| |
Collapse
|
284
|
Ledo AM, Senra A, Rilo-Alvarez H, Borrajo E, Vidal A, Alonso MJ, Garcia-Fuentes M. mRNA-activated matrices encoding transcription factors as primers of cell differentiation in tissue engineering. Biomaterials 2020; 247:120016. [PMID: 32272302 DOI: 10.1016/j.biomaterials.2020.120016] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 03/19/2020] [Accepted: 03/27/2020] [Indexed: 12/16/2022]
Abstract
Gene-activated matrices (GAMs) encoding pivotal transcription factors (TFs) represent a powerful tool to direct stem cell specification for tissue engineering applications. However, current TF-based GAMs activated with pDNA, are challenged by their low transfection efficiency and delayed transgene expression. Here, we report a GAM technology activated with mRNAs encoding TFs SOX9 (cartilage) and MYOD (muscle). We find that these mRNA-GAMs induce a higher and faster TF expression compared to pDNA-GAMs, especially in the case of RNase resistant mRNA sequences. This potent TF expression was translated into a high synthesis of cartilage- and muscle-specific markers, and ultimately, into successful tissue specification in vitro. Additionally, we show that the expression of tissue-specific markers can be further modulated by altering the properties of the mRNA-GAM environment. These results highlight the value of this GAM technology for priming cell lineage specification, a key centerpiece for future tissue engineering devices.
Collapse
Affiliation(s)
- Adriana M Ledo
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, IDIS Research Institute, CiMUS Research Institute, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Ana Senra
- Histology Unit, CiMUS Research Institute, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Héctor Rilo-Alvarez
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, IDIS Research Institute, CiMUS Research Institute, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Erea Borrajo
- Department of Physiology, IDIS Research Institute, CiMUS Research Institute, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Anxo Vidal
- Department of Physiology, IDIS Research Institute, CiMUS Research Institute, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Maria J Alonso
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, IDIS Research Institute, CiMUS Research Institute, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Marcos Garcia-Fuentes
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, IDIS Research Institute, CiMUS Research Institute, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain.
| |
Collapse
|
285
|
Venkatesan JK, Meng W, Rey-Rico A, Schmitt G, Speicher-Mentges S, Falentin-Daudré C, Leroux A, Madry H, Migonney V, Cucchiarini M. Enhanced Chondrogenic Differentiation Activities in Human Bone Marrow Aspirates via sox9 Overexpression Mediated by pNaSS-Grafted PCL Film-Guided rAAV Gene Transfer. Pharmaceutics 2020; 12:pharmaceutics12030280. [PMID: 32245159 PMCID: PMC7151167 DOI: 10.3390/pharmaceutics12030280] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/13/2020] [Accepted: 03/19/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The delivery of therapeutic genes in sites of articular cartilage lesions using non-invasive, scaffold-guided gene therapy procedures is a promising approach to stimulate cartilage repair while protecting the cargos from detrimental immune responses, particularly when targeting chondroreparative bone marrow-derived mesenchymal stromal cells in a natural microenvironment like marrow aspirates. METHODS Here, we evaluated the benefits of providing a sequence for the cartilage-specific sex-determining region Y-type high-mobility group box 9 (SOX9) transcription factor to human marrow aspirates via recombinant adeno-associated virus (rAAV) vectors delivered by poly(ε-caprolactone) (PCL) films functionalized via grafting with poly(sodium styrene sulfonate) (pNaSS) to enhance the marrow chondrogenic potential over time. RESULTS Effective sox9 overexpression was observed in aspirates treated with pNaSS-grafted or ungrafted PCL films coated with the candidate rAAV-FLAG-hsox9 (FLAG-tagged rAAV vector carrying a human sox9 gene sequence) vector for at least 21 days relative to other conditions (pNaSS-grafted and ungrafted PCL films without vector coating). Overexpression of sox9 via rAAV sox9/pNaSS-grafted or ungrafted PCL films led to increased biological and chondrogenic differentiation activities (matrix deposition) in the aspirates while containing premature osteogenesis and hypertrophy without impacting cell proliferation, with more potent effects noted when using pNaSS-grafted films. CONCLUSIONS These findings show the benefits of targeting patients' bone marrow via PCL film-guided therapeutic rAAV (sox9) delivery as an off-the-shelf system for future strategies to enhance cartilage repair in translational applications.
Collapse
Affiliation(s)
- Jagadeesh K. Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, D-66421 Homburg/Saar, Germany; (J.K.V.); (W.M.); (A.R.-R.); (G.S.); (S.S.-M.); (H.M.)
| | - Weikun Meng
- Center of Experimental Orthopaedics, Saarland University Medical Center, D-66421 Homburg/Saar, Germany; (J.K.V.); (W.M.); (A.R.-R.); (G.S.); (S.S.-M.); (H.M.)
| | - Ana Rey-Rico
- Center of Experimental Orthopaedics, Saarland University Medical Center, D-66421 Homburg/Saar, Germany; (J.K.V.); (W.M.); (A.R.-R.); (G.S.); (S.S.-M.); (H.M.)
| | - Gertrud Schmitt
- Center of Experimental Orthopaedics, Saarland University Medical Center, D-66421 Homburg/Saar, Germany; (J.K.V.); (W.M.); (A.R.-R.); (G.S.); (S.S.-M.); (H.M.)
| | - Susanne Speicher-Mentges
- Center of Experimental Orthopaedics, Saarland University Medical Center, D-66421 Homburg/Saar, Germany; (J.K.V.); (W.M.); (A.R.-R.); (G.S.); (S.S.-M.); (H.M.)
| | - Céline Falentin-Daudré
- LBPS/CSPBAT UMR CNRS 7244, Université Sorbonne Paris Nord, F-93430 Villetaneuse, France; (C.F.-D.); (A.L.); (V.M.)
| | - Amélie Leroux
- LBPS/CSPBAT UMR CNRS 7244, Université Sorbonne Paris Nord, F-93430 Villetaneuse, France; (C.F.-D.); (A.L.); (V.M.)
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, D-66421 Homburg/Saar, Germany; (J.K.V.); (W.M.); (A.R.-R.); (G.S.); (S.S.-M.); (H.M.)
- Department of Orthopaedic Surgery, Saarland University Medical Center, D-66421 Homburg/Saar, Germany
| | - Véronique Migonney
- LBPS/CSPBAT UMR CNRS 7244, Université Sorbonne Paris Nord, F-93430 Villetaneuse, France; (C.F.-D.); (A.L.); (V.M.)
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, D-66421 Homburg/Saar, Germany; (J.K.V.); (W.M.); (A.R.-R.); (G.S.); (S.S.-M.); (H.M.)
- Correspondence: ; Tel.: +49-6841-1624-987; Fax: +49-6841-1624-988
| |
Collapse
|
286
|
Ibaraki K, Hayashi S, Kanzaki N, Hashimoto S, Kihara S, Haneda M, Takeuchi K, Niikura T, Kuroda R. Deletion of p21 expression accelerates cartilage tissue repair via chondrocyte proliferation. Mol Med Rep 2020; 21:2236-2242. [PMID: 32186772 DOI: 10.3892/mmr.2020.11028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 02/03/2020] [Indexed: 11/05/2022] Open
Abstract
Articular cartilage tissue has a poor healing potential, and when subjected to traumatic damage this tissue undergoes cartilage degeneration and osteoarthritis. The association between the regulation of cell cycle checkpoints and tissue regeneration has been previously investigated, and p21 was initially identified as a potent inhibitor of cell cycle progression. However, the effects of p21 defects on damaged tissue remain controversial. Therefore, the aim of the present study was to evaluate the effects of p21 deficiency on cartilage repair. A mouse model of articular cartilage repair was generated by inducing a patellar groove scratch in 8‑week‑old p21‑knockout (KO) mice and C57Bl/6 wild‑type (WT) mice. Mice were sacrificed at 4 and 8 weeks post‑surgery. The present study also investigated the effect of p21 deficiency on cartilage differentiation in ATDC5 cells in vitro. Safranin O staining results indicated that cartilage repair initially occurred in p21 KO mice. In addition, immunohistochemical analysis demonstrated that p21 KO upregulated proliferating cell nuclear antigen and increased cell proliferation. However, type II collagen and Sox9 expression levels remained unchanged in p21 KO and WT mice. Moreover, it was identified that p21 downregulation did not affect Sox9 and type II collagen expression levels in vitro. Furthermore, p21 deficiency promoted healing of articular cartilage damage, which was associated with cell proliferation in vivo, and increased chondrocyte proliferation but not differentiation in vitro. Therefore, the present results suggested that p21 does not affect Sox9 or type II collagen expression levels during cartilage differentiation in the repair of cartilage defects.
Collapse
Affiliation(s)
- Kazuyuki Ibaraki
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Shinya Hayashi
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Noriyuki Kanzaki
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Shingo Hashimoto
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Shinsuke Kihara
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Masahiko Haneda
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Kazuhiro Takeuchi
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Takahiro Niikura
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Ryosuke Kuroda
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| |
Collapse
|
287
|
Xu G, Liu Y, Zhang C, Zhou Y, Hou S, Tang J, Li Z. Temporal and spatial expression of Sox9, Pax1, TGF-β1 and type I and II collagen in human intervertebral disc development. Neurochirurgie 2020; 66:168-173. [PMID: 32201238 DOI: 10.1016/j.neuchi.2019.12.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 11/06/2019] [Accepted: 12/08/2019] [Indexed: 10/24/2022]
Abstract
PURPOSE An accurate understanding of cellular biochemical changes in human intervertebral disc (IVD)s and the corresponding mechanisms during the developmental process still remain unknown and important for investigating the function of critical factors in normal IVD development as well as ascertaining the therapeutic targets for the IVD degeneration. METHODS Under ethical conditions, human fetal cervical IVDs at 4, 5, and 6 months of pregnancy were collected at abortion surgery. Normal adult human C3-C7 cervical IVDs were taken from cadaveric donors. Sox9, Pax1, TGF-β1 and type I/II collagen protein and RNA were detected. The number of positive cells was counted to calculate the optical density value for each factor. RESULTS Sox9, Pax1, and TGF-β1 expression in the IVD was remarkably reduced with the developmental stage. The location of high expression of Sox9, Pax1, and TGF-β1 changed with the developmental stage, and migrated from the nucleus pulposus to the annulus fibrosus and endplate. Higher Sox9, Pax1, and TGF-β1 expression was finally observed around the sclerotome of the vertebral body. The anabolism of type I/II collagens is significantly increased in the IVD in the mid-trimester fetus. CONCLUSIONS Sox9, Pax1 and TGF-β1 participate in the developmental process of the human IVD and vertebral body. However, these factors show a separate expression of mRNA and protein, suggesting that they are expressed in the strict time and spatial order.
Collapse
Affiliation(s)
- G Xu
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Diseases Liaoning Province, Department of Orthopaedics, First Affiliated Hospital of Dalian Medical University, 116011 Dalian, People's Republic of China
| | - Y Liu
- Department of Orthopedics, First Affiliated Hospital of PLA General Hospital, 100048 Beijing, People's Republic of China
| | - C Zhang
- Department of Orthopedics, First Affiliated Hospital of PLA General Hospital, 100048 Beijing, People's Republic of China
| | - Y Zhou
- Department of Orthopedics, First Affiliated Hospital of PLA General Hospital, 100048 Beijing, People's Republic of China
| | - S Hou
- Department of Orthopedics, First Affiliated Hospital of PLA General Hospital, 100048 Beijing, People's Republic of China
| | - J Tang
- Department of Orthopedics, First Affiliated Hospital of PLA General Hospital, 100048 Beijing, People's Republic of China.
| | - Z Li
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Diseases Liaoning Province, Department of Orthopaedics, First Affiliated Hospital of Dalian Medical University, 116011 Dalian, People's Republic of China.
| |
Collapse
|
288
|
Yan H, Hales BF. Effects of Organophosphate Ester Flame Retardants on Endochondral Ossification in Ex Vivo Murine Limb Bud Cultures. Toxicol Sci 2020; 168:420-429. [PMID: 30561715 DOI: 10.1093/toxsci/kfy301] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Phasing out the usage of polybrominated diphenyl ether (PBDE) flame retardants (FRs) in consumer products led to their widespread replacement with organophosphate ester (OPE) FRs, despite scarce safety data. PBDE exposures were associated with the suppression of endochondral ossification but little is known about the effects of OPEs on bones. Here, we used a novel ex vivo murine limb bud culture system to compare the effects of 2,2',4,4'-tetrabromodiphenyl ether (BDE-47) with those of several OPEs. Gestation day 13 embryos were collected from transgenic CD1 mice expressing fluorescent markers for the major stages of endochondral ossification: COL2A1-ECFP (chondrogenesis), COL10A1-mCherry (early osteogenesis), and COL1A1-YFP (late osteogenesis). Limbs were excised and cultured for 6 days in the presence of vehicle, BDE-47, or an OPE FR: triphenyl phosphate (TPHP), tert-butylphenyl diphenyl phosphate (BPDP), tris(methylphenyl) phosphate (TMPP), or isopropylated triphenyl phosphate (IPPP). BDE-47 (50 μM) decreased the extent of chondrogenesis in the digits and COL1A1-YFP expression in the radius and ulna relative to control. In comparison, concentrations of ≥1 μM of all 4 OPEs limited chondrogenesis; osteogenesis (both COL10A1-mCherry and COL1A1-YFP fluorescence) was markedly inhibited at concentrations ≥3 μM. The expression of Sox9, the master regulator of chondrogenesis, was altered by BDE-47, TPHP, and BPDP. BDE-47 exposure had minimal impact on the expression of Runx2 and Sp7, which drive osteogenesis, whereas TPHP and BPDP both suppressed the expression of these transcription factors. These data suggest that OPE FRs may be more detrimental to bone formation than their brominated predecessors.
Collapse
Affiliation(s)
- Han Yan
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Barbara F Hales
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada
| |
Collapse
|
289
|
Wise CA, Sepich D, Ushiki A, Khanshour AM, Kidane YH, Makki N, Gurnett CA, Gray RS, Rios JJ, Ahituv N, Solnica-Krezel L. The cartilage matrisome in adolescent idiopathic scoliosis. Bone Res 2020; 8:13. [PMID: 32195011 PMCID: PMC7062733 DOI: 10.1038/s41413-020-0089-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 01/27/2020] [Indexed: 12/13/2022] Open
Abstract
The human spinal column is a dynamic, segmented, bony, and cartilaginous structure that protects the neurologic system and simultaneously provides balance and flexibility. Children with developmental disorders that affect the patterning or shape of the spine can be at risk of neurologic and other physiologic dysfunctions. The most common developmental disorder of the spine is scoliosis, a lateral deformity in the shape of the spinal column. Scoliosis may be part of the clinical spectrum that is observed in many developmental disorders, but typically presents as an isolated symptom in otherwise healthy adolescent children. Adolescent idiopathic scoliosis (AIS) has defied understanding in part due to its genetic complexity. Breakthroughs have come from recent genome-wide association studies (GWAS) and next generation sequencing (NGS) of human AIS cohorts, as well as investigations of animal models. These studies have identified genetic associations with determinants of cartilage biogenesis and development of the intervertebral disc (IVD). Current evidence suggests that a fraction of AIS cases may arise from variation in factors involved in the structural integrity and homeostasis of the cartilaginous extracellular matrix (ECM). Here, we review the development of the spine and spinal cartilages, the composition of the cartilage ECM, the so-called "matrisome" and its functions, and the players involved in the genetic architecture of AIS. We also propose a molecular model by which the cartilage matrisome of the IVD contributes to AIS susceptibility.
Collapse
Affiliation(s)
- Carol A. Wise
- Center for Pediatric Bone Biology and Translational Research, Texas Scottish Rite Hospital for Children, 2222 Welborn St., Dallas, TX 75219 USA
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75235 USA
- Departments of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75235 USA
- Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75235 USA
| | - Diane Sepich
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Aki Ushiki
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94158 USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94158 USA
| | - Anas M. Khanshour
- Center for Pediatric Bone Biology and Translational Research, Texas Scottish Rite Hospital for Children, 2222 Welborn St., Dallas, TX 75219 USA
| | - Yared H. Kidane
- Center for Pediatric Bone Biology and Translational Research, Texas Scottish Rite Hospital for Children, 2222 Welborn St., Dallas, TX 75219 USA
| | - Nadja Makki
- Department of Anatomy and Cell Biology, University of Florida, College of Medicine, Gainesville, FL 32610 USA
| | - Christina A. Gurnett
- Departments of Neurology, Washington University School of Medicine, St Louis, MO 63110 USA
- Pediatrics, Washington University School of Medicine, St Louis, MO 63110 USA
- Orthopaedic Surgery, Washington University School of Medicine, St Louis, MO 63110 USA
| | - Ryan S. Gray
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin Dell Medical School, Austin, TX 78723 USA
| | - Jonathan J. Rios
- Center for Pediatric Bone Biology and Translational Research, Texas Scottish Rite Hospital for Children, 2222 Welborn St., Dallas, TX 75219 USA
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75235 USA
- Departments of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75235 USA
- Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75235 USA
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94158 USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94158 USA
| | - Lila Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
| |
Collapse
|
290
|
Transcriptional analysis of scar-free wound healing during early stages of tail regeneration in the green anole lizard, Anolis carolinensis. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.regen.2019.100025] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
291
|
Nakamichi R, Asahara H. The transcription factors regulating intervertebral disc development. JOR Spine 2020; 3:e1081. [PMID: 32211592 PMCID: PMC7084052 DOI: 10.1002/jsp2.1081] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 01/22/2020] [Accepted: 02/01/2020] [Indexed: 01/06/2023] Open
Abstract
Damage to the intervertebral discs (IVDs) occurs due to aging or excessive mechanical stress, causing a series of IVD-related degenerative diseases, such as spinal disc herniation and spondylosis. These IVD-related diseases are difficult to cure, partially because the regeneration ability of IVDs is not sufficient. As a novel strategy for treatment of IVD-related diseases, mesenchymal stem cell transplantation to the damaged discs has been reported in animal studies. To further develop and improve this approach, it is necessary to gain a better understanding of the molecular network regulating IVD development by critical transcription factors. Recent findings reveal that during IVD development, nucleus pulposus and annuls fibrosus differentiation is coordinated by a series of transcription factors, such as Mkx, Pax1, 9, Shh, Foxa1, 2, T-Brachyury, and Sox5, 6, 9. The combination of mesenchymal stem cell transplantation with the regulation of these molecules may provide a novel strategy for treatment of degenerative disc diseases.
Collapse
Affiliation(s)
- Ryo Nakamichi
- Department of Molecular and Experimental MedicineThe Scripps Research InstituteLa JollaCalifornia
- Department of Orthopaedic SurgeryOkayama University Graduate School of Medicine, Dentistry, and Pharmaceutical SciencesOkayamaJapan
| | - Hiroshi Asahara
- Department of Molecular and Experimental MedicineThe Scripps Research InstituteLa JollaCalifornia
- Department of Systems BiomedicineTokyo Medical and Dental UniversityTokyoJapan
| |
Collapse
|
292
|
Pathomechanisms of Posttraumatic Osteoarthritis: Chondrocyte Behavior and Fate in a Precarious Environment. Int J Mol Sci 2020; 21:ijms21051560. [PMID: 32106481 PMCID: PMC7084733 DOI: 10.3390/ijms21051560] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/18/2020] [Accepted: 02/21/2020] [Indexed: 02/07/2023] Open
Abstract
Traumatic injuries of the knee joint result in a wide variety of pathomechanisms, which contribute to the development of so-called posttraumatic osteoarthritis (PTOA). These pathogenetic processes include oxidative stress, excessive expression of catabolic enzymes, release of damage-associated molecular patterns (DAMPs), and synovial inflammation. The present review focuses on the underlying pathomechanisms of PTOA and in particular the behavior and fate of the surviving chondrocytes, comprising chondrocyte metabolism, regulated cell death, and phenotypical changes comprising hypertrophy and senescence. Moreover, possible therapeutic strategies, such as chondroanabolic stimulation, anti-oxidative and anti-inflammatory treatment, as well as novel therapeutic targets are discussed.
Collapse
|
293
|
Anderson-Baron M, Kunze M, Mulet-Sierra A, Adesida AB. Effect of cell seeding density on matrix-forming capacity of meniscus fibrochondrocytes and nasal chondrocytes in meniscus tissue engineering. FASEB J 2020; 34:5538-5551. [PMID: 32090374 DOI: 10.1096/fj.201902559r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 02/04/2020] [Accepted: 02/13/2020] [Indexed: 12/11/2022]
Abstract
The presence of intact menisci is imperative for the proper function of the knee joint. Meniscus injuries are often treated by the surgical removal of the damaged tissue, which increases the likelihood of post-traumatic osteoarthritis. Tissue engineering holds great promise in producing viable engineered meniscal tissue for implantation using the patient's own cells; however, the cell source for producing the engineered tissue is unclear. Nasal chondrocytes (NC) possess many attractive features for engineering meniscus. However, in order to validate the use of NC for engineering meniscus fibrocartilage, a thorough comparison of NC and meniscus fibrochondrocytes (MFC) must be considered. Our study presents an analysis of the relative features of NC and MFC and their respective chondrogenic potential in a pellet culture model. We showed considerable differences in the cartilage tissue formed by the two different cell types. Our data showed that NC were more proliferative in culture, deposited more extracellular matrix, and showed higher expression of chondrogenic genes than MFC. Overall, our data suggest that NC produce superior cartilage tissue to MFC in a pellet culture model. In addition, NCs produce higher quality cartilage tissue at higher cell seeding densities during cell expansion.
Collapse
Affiliation(s)
- Matthew Anderson-Baron
- Department of Surgery, Divisions of Orthopaedic Surgery and Surgical Research, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada.,Laboratory of Stem Cell Biology and Orthopaedic Tissue Engineering, 3-021 Li Ka Shing Centre for Health Research Innovation, Edmonton, AB, Canada
| | - Melanie Kunze
- Department of Surgery, Divisions of Orthopaedic Surgery and Surgical Research, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada.,Laboratory of Stem Cell Biology and Orthopaedic Tissue Engineering, 3-021 Li Ka Shing Centre for Health Research Innovation, Edmonton, AB, Canada
| | - Aillette Mulet-Sierra
- Department of Surgery, Divisions of Orthopaedic Surgery and Surgical Research, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada.,Laboratory of Stem Cell Biology and Orthopaedic Tissue Engineering, 3-021 Li Ka Shing Centre for Health Research Innovation, Edmonton, AB, Canada
| | - Adetola B Adesida
- Department of Surgery, Divisions of Orthopaedic Surgery and Surgical Research, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada.,Laboratory of Stem Cell Biology and Orthopaedic Tissue Engineering, 3-021 Li Ka Shing Centre for Health Research Innovation, Edmonton, AB, Canada
| |
Collapse
|
294
|
Yilmaz EN, Zeugolis DI. Electrospun Polymers in Cartilage Engineering-State of Play. Front Bioeng Biotechnol 2020; 8:77. [PMID: 32133352 PMCID: PMC7039817 DOI: 10.3389/fbioe.2020.00077] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 01/29/2020] [Indexed: 12/17/2022] Open
Abstract
Articular cartilage defects remain a clinical challenge. Articular cartilage defects progress to osteoarthritis, which negatively (e.g., remarkable pain, decreased mobility, distress) affects millions of people worldwide and is associated with excessive healthcare costs. Surgical procedures and cell-based therapies have failed to deliver a functional therapy. To this end, tissue engineering therapies provide a promise to deliver a functional cartilage substitute. Among the various scaffold fabrication technologies available, electrospinning is continuously gaining pace, as it can produce nano- to micro- fibrous scaffolds that imitate architectural features of native extracellular matrix supramolecular assemblies and can deliver variable cell populations and bioactive molecules. Herein, we comprehensively review advancements and shortfalls of various electrospun scaffolds in cartilage engineering.
Collapse
Affiliation(s)
- Elif Nur Yilmaz
- Regenerative, Modular & Developmental Engineering Laboratory, National University of Ireland Galway, Galway, Ireland.,Science Foundation Ireland, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory, National University of Ireland Galway, Galway, Ireland.,Science Foundation Ireland, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
295
|
Weissenberger M, Weissenberger MH, Gilbert F, Groll J, Evans CH, Steinert AF. Reduced hypertrophy in vitro after chondrogenic differentiation of adult human mesenchymal stem cells following adenoviral SOX9 gene delivery. BMC Musculoskelet Disord 2020; 21:109. [PMID: 32066427 PMCID: PMC7026978 DOI: 10.1186/s12891-020-3137-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 02/12/2020] [Indexed: 01/03/2023] Open
Abstract
Background Mesenchymal stem cell (MSC) based-treatments of cartilage injury are promising but impaired by high levels of hypertrophy after chondrogenic induction with several bone morphogenetic protein superfamily members (BMPs). As an alternative, this study investigates the chondrogenic induction of MSCs via adenoviral gene-delivery of the transcription factor SOX9 alone or in combination with other inducers, and comparatively explores the levels of hypertrophy and end stage differentiation in a pellet culture system in vitro. Methods First generation adenoviral vectors encoding SOX9, TGFB1 or IGF1 were used alone or in combination to transduce human bone marrow-derived MSCs at 5 × 102 infectious particles/cell. Thereafter cells were placed in aggregates and maintained for three weeks in chondrogenic medium. Transgene expression was determined at the protein level (ELISA/Western blot), and aggregates were analysed histologically, immunohistochemically, biochemically and by RT-PCR for chondrogenesis and hypertrophy. Results SOX9 cDNA was superior to that encoding TGFB1, the typical gold standard, as an inducer of chondrogenesis in primary MSCs as evidenced by improved lacuna formation, proteoglycan and collagen type II staining, increased levels of GAG synthesis, and expression of mRNAs associated with chondrogenesis. Moreover, SOX9 modified aggregates showed a markedly lower tendency to progress towards hypertrophy, as judged by expression of the hypertrophy markers alkaline phosphatase, and collagen type X at the mRNA and protein levels. Conclusion Adenoviral SOX9 gene transfer induces chondrogenic differentiation of human primary MSCs in pellet culture more effectively than TGFB1 gene transfer with lower levels of chondrocyte hypertrophy after 3 weeks of in vitro culture. Such technology might enable the formation of more stable hyaline cartilage repair tissues in vivo.
Collapse
Affiliation(s)
- M Weissenberger
- Department of Orthopaedic Surgery, König-Ludwig-Haus, Center for Musculoskeletal Research, Julius-Maximilians-University, Brettreichstrasse 11, D-97074, Würzburg, Germany.
| | - M H Weissenberger
- Department of Orthopaedic Surgery, König-Ludwig-Haus, Center for Musculoskeletal Research, Julius-Maximilians-University, Brettreichstrasse 11, D-97074, Würzburg, Germany.,Department of Pathology, Caritas-Hospital, Bad Mergentheim, Germany
| | - F Gilbert
- Department of Orthopaedic Surgery, König-Ludwig-Haus, Center for Musculoskeletal Research, Julius-Maximilians-University, Brettreichstrasse 11, D-97074, Würzburg, Germany.,Department of Orthopaedic Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Würzburg, Würzburg, Germany
| | - J Groll
- Department of Functional Materials in Medicine and Dentistry, Julius-Maximilians-University, Würzburg, Germany
| | - C H Evans
- Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, MN, USA
| | - A F Steinert
- Department of Orthopaedic Surgery, König-Ludwig-Haus, Center for Musculoskeletal Research, Julius-Maximilians-University, Brettreichstrasse 11, D-97074, Würzburg, Germany.,Present address: Department of Orthopaedic, Trauma, Shoulder and Arthroplasty Surgery, Rhön-Klinikum Campus Bad Neustadt, Bad Neustadt a.d. Saale, Germany
| |
Collapse
|
296
|
Nwadozi E, Rudnicki M, Haas TL. Metabolic Coordination of Pericyte Phenotypes: Therapeutic Implications. Front Cell Dev Biol 2020; 8:77. [PMID: 32117997 PMCID: PMC7033550 DOI: 10.3389/fcell.2020.00077] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/29/2020] [Indexed: 12/15/2022] Open
Abstract
Pericytes are mural vascular cells found predominantly on the abluminal wall of capillaries, where they contribute to the maintenance of capillary structural integrity and vascular permeability. Generally quiescent cells in the adult, pericyte activation and proliferation occur during both physiological and pathological vascular and tissue remodeling. A considerable body of research indicates that pericytes possess attributes of a multipotent adult stem cell, as they are capable of self-renewal as well as commitment and differentiation into multiple lineages. However, pericytes also display phenotypic heterogeneity and recent studies indicate that lineage potential differs between pericyte subpopulations. While numerous microenvironmental cues and cell signaling pathways are known to regulate pericyte functions, the roles that metabolic pathways play in pericyte quiescence, self-renewal or differentiation have been given limited consideration to date. This review will summarize existing data regarding pericyte metabolism and will discuss the coupling of signal pathways to shifts in metabolic pathway preferences that ultimately regulate pericyte quiescence, self-renewal and trans-differentiation. The association between dysregulated metabolic processes and development of pericyte pathologies will be highlighted. Despite ongoing debate regarding pericyte classification and their functional capacity for trans-differentiation in vivo, pericytes are increasingly exploited as a cell therapy tool to promote tissue healing and regeneration. Ultimately, the efficacy of therapeutic approaches hinges on the capacity to effectively control/optimize the fate of the implanted pericytes. Thus, we will identify knowledge gaps that need to be addressed to more effectively harness the opportunity for therapeutic manipulation of pericytes to control pathological outcomes in tissue remodeling.
Collapse
Affiliation(s)
| | | | - Tara L. Haas
- School of Kinesiology and Health Science, Angiogenesis Research Group and Muscle Health Research Centre, York University, Toronto, ON, Canada
| |
Collapse
|
297
|
Generation of a Quantitative Luciferase Reporter for Sox9 SUMOylation. Int J Mol Sci 2020; 21:ijms21041274. [PMID: 32070068 PMCID: PMC7072981 DOI: 10.3390/ijms21041274] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 01/30/2020] [Accepted: 02/11/2020] [Indexed: 01/11/2023] Open
Abstract
Sox9 is a master transcription factor for chondrogenesis, which is essential for chondrocyte proliferation, differentiation, and maintenance. Sox9 activity is regulated by multiple layers, including post-translational modifications, such as SUMOylation. A detection method for visualizing the SUMOylation in live cells is required to fully understand the role of Sox9 SUMOylation. In this study, we generated a quantitative reporter for Sox9 SUMOylation that is based on the NanoBiT system. The simultaneous expression of Sox9 and SUMO1 constructs that are conjugated with NanoBiT fragments in HEK293T cells induced luciferase activity in SUMOylation target residue of Sox9-dependent manner. Furthermore, the reporter signal could be detected from both cell lysates and live cells. The signal level of our reporter responded to the co-expression of SUMOylation or deSUMOylation enzymes by several fold, showing dynamic potency of the reporter. The reporter was active in multiple cell types, including ATDC5 cells, which have chondrogenic potential. Finally, using this reporter, we revealed a extracellular signal conditions that can increase the amount of SUMOylated Sox9. In summary, we generated a novel reporter that was capable of quantitatively visualizing the Sox9-SUMOylation level in live cells. This reporter will be useful for understanding the dynamism of Sox9 regulation during chondrogenesis.
Collapse
|
298
|
Zhi Z, Zhang C, Kang J, Wang Y, Liu J, Wu F, Xu G. The therapeutic effect of bone marrow-derived mesenchymal stem cells on osteoarthritis is improved by the activation of the KDM6A/SOX9 signaling pathway caused by exposure to hypoxia. J Cell Physiol 2020; 235:7173-7182. [PMID: 32020624 DOI: 10.1002/jcp.29615] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 01/09/2020] [Indexed: 12/27/2022]
Abstract
Abnormal expression of KDM6A and SOX9 is a key factor in the pathogenesis of osteoarthritis (OA). Cellular treatments of OA with articular cartilage chondrocytes (ACCs) and bone marrow mesenchymal stem cells (BMSCs) are promising, but their underlying mechanisms remain to be explored. The pellet size, weight and sulfated glycosaminoglycan/DNA content of ACCs were measured to evaluate the effect of BMSCs on the chondrogenic differentiation of SCCs. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was used to analyze the proliferation of ACCs cultured along or cocultured with BMSCs. Quantitative polymerase chain reaction (qPCR) was performed to evaluate the messenger RNA expression of KDM6A, SOX9, type2 collagen, and Aggrecan in ACCs and OA rats. Western blot and immunohistochemistry were performed to analyze the expression of KDM6A and SOX9 proteins. Bisulfite sequencing PCR was performed to assess the DNA methylation level of the SOX9 promoter. Flow cytometry was used to evaluate the apoptotic status of ACCs. The chondrogenic differentiation of ACCs was significantly enhanced by coculturing with BMSCs, especially under a hypoxic condition. The expression of KDM6A, SOX9, type2 collagen, and Aggrecan was remarkably elevated in ACCs cocultured with BMSCs. Also, the DNA methylation of SOX9 promoter was decreased in ACCs cocultured with BMSCs, along with notably reduced apoptosis. Moreover, ACCs cocultured with BMSCs could repair cartilage lesions and prevent the abnormal expression of KDM6A, SOX9, type2 collagen, and Aggrecan in OA rats. In this study, we cocultured ACCs with BMSCs and used them to treat OA rats. Our findings presented a mechanistic basis for explaining the therapeutic effect of BMSCs on OA treatment.
Collapse
Affiliation(s)
- Zhongzheng Zhi
- Department of Orthopedics, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Chenglin Zhang
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jian Kang
- Department of Orthopedics, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Yingjie Wang
- Department of Orthopedics, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Jingdong Liu
- Department of Orthopedics, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Furong Wu
- Shanghai Clinical Research Center for Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guanghui Xu
- Department of Orthopedics, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
299
|
De la Vega RE, Scheu M, Brown LA, Evans CH, Ferreira E, Porter RM. Specific, Sensitive, and Stable Reporting of Human Mesenchymal Stromal Cell Chondrogenesis. Tissue Eng Part C Methods 2020; 25:176-190. [PMID: 30727864 DOI: 10.1089/ten.tec.2018.0295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
IMPACT STATEMENT The promoter characterized in this study has been made accessible as a resource for the skeletal tissue engineering and regenerative medicine community. When combined with suitable reporter vectors, the resulting tools can be used for noninvasive and/or high-throughput screening of test conditions for stimulating chondrogenesis by candidate stem/progenitor cells. As demonstrated in this study, they can also be used with small animal imaging platforms to monitor the chondrogenic activity of implanted progenitors within orthotopic models of bone and cartilage repair.
Collapse
Affiliation(s)
- Rodolfo E De la Vega
- 1 Department of Orthopaedic Surgery, Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center, Boston, Massachusetts.,2 Department of Orthopaedic Surgery, Harvard Medical School, Boston, Massachusetts
| | - Maximiliano Scheu
- 1 Department of Orthopaedic Surgery, Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center, Boston, Massachusetts.,2 Department of Orthopaedic Surgery, Harvard Medical School, Boston, Massachusetts.,3 Department of Orthopaedic Surgery, Clínica Alemana de Santiago, Universidad del Desarrollo, Vitacura, Chile
| | - Lennart A Brown
- 1 Department of Orthopaedic Surgery, Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center, Boston, Massachusetts.,2 Department of Orthopaedic Surgery, Harvard Medical School, Boston, Massachusetts
| | - Christopher H Evans
- 1 Department of Orthopaedic Surgery, Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center, Boston, Massachusetts.,2 Department of Orthopaedic Surgery, Harvard Medical School, Boston, Massachusetts
| | - Elisabeth Ferreira
- 1 Department of Orthopaedic Surgery, Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center, Boston, Massachusetts.,2 Department of Orthopaedic Surgery, Harvard Medical School, Boston, Massachusetts
| | - Ryan M Porter
- 1 Department of Orthopaedic Surgery, Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center, Boston, Massachusetts.,2 Department of Orthopaedic Surgery, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
300
|
Sebastian A, Murugesh DK, Mendez ME, Hum NR, Rios-Arce ND, McCool JL, Christiansen BA, Loots GG. Global Gene Expression Analysis Identifies Age-Related Differences in Knee Joint Transcriptome during the Development of Post-Traumatic Osteoarthritis in Mice. Int J Mol Sci 2020; 21:ijms21010364. [PMID: 31935848 PMCID: PMC6982134 DOI: 10.3390/ijms21010364] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 12/29/2019] [Accepted: 12/29/2019] [Indexed: 12/15/2022] Open
Abstract
Aging and injury are two major risk factors for osteoarthritis (OA). Yet, very little is known about how aging and injury interact and contribute to OA pathogenesis. In the present study, we examined age- and injury-related molecular changes in mouse knee joints that could contribute to OA. Using RNA-seq, first we profiled the knee joint transcriptome of 10-week-old, 62-week-old, and 95-week-old mice and found that the expression of several inflammatory-response related genes increased as a result of aging, whereas the expression of several genes involved in cartilage metabolism decreased with age. To determine how aging impacts post-traumatic arthritis (PTOA) development, the right knee joints of 10-week-old and 62-week-old mice were injured using a non-invasive tibial compression injury model and injury-induced structural and molecular changes were assessed. At six-week post-injury, 62-week-old mice displayed significantly more cartilage degeneration and osteophyte formation compared with young mice. Although both age groups elicited similar transcriptional responses to injury, 62-week-old mice had higher activation of inflammatory cytokines than 10-week-old mice, whereas cartilage/bone metabolism genes had higher expression in 10-week-old mice, suggesting that the differential expression of these genes might contribute to the differences in PTOA severity observed between these age groups.
Collapse
Affiliation(s)
- Aimy Sebastian
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories, Livermore, CA 94550, USA; (A.S.); (D.K.M.); (M.E.M.); (N.R.H.); (N.D.R.-A.); (J.L.M.)
| | - Deepa K. Murugesh
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories, Livermore, CA 94550, USA; (A.S.); (D.K.M.); (M.E.M.); (N.R.H.); (N.D.R.-A.); (J.L.M.)
| | - Melanie E. Mendez
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories, Livermore, CA 94550, USA; (A.S.); (D.K.M.); (M.E.M.); (N.R.H.); (N.D.R.-A.); (J.L.M.)
- Molecular and Cell Biology, School of Natural Sciences, UC Merced, Merced, CA 95343, USA
| | - Nicholas R. Hum
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories, Livermore, CA 94550, USA; (A.S.); (D.K.M.); (M.E.M.); (N.R.H.); (N.D.R.-A.); (J.L.M.)
- Molecular and Cell Biology, School of Natural Sciences, UC Merced, Merced, CA 95343, USA
| | - Naiomy D. Rios-Arce
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories, Livermore, CA 94550, USA; (A.S.); (D.K.M.); (M.E.M.); (N.R.H.); (N.D.R.-A.); (J.L.M.)
| | - Jillian L. McCool
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories, Livermore, CA 94550, USA; (A.S.); (D.K.M.); (M.E.M.); (N.R.H.); (N.D.R.-A.); (J.L.M.)
- Molecular and Cell Biology, School of Natural Sciences, UC Merced, Merced, CA 95343, USA
| | | | - Gabriela G. Loots
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories, Livermore, CA 94550, USA; (A.S.); (D.K.M.); (M.E.M.); (N.R.H.); (N.D.R.-A.); (J.L.M.)
- Molecular and Cell Biology, School of Natural Sciences, UC Merced, Merced, CA 95343, USA
- Correspondence: ; Tel.: +1-925-423-0923
| |
Collapse
|