151
|
Scialla S, Gullotta F, Izzo D, Palazzo B, Scalera F, Martin I, Sannino A, Gervaso F. Genipin-crosslinked collagen scaffolds inducing chondrogenesis: a mechanical and biological characterization. J Biomed Mater Res A 2022; 110:1372-1385. [PMID: 35262240 DOI: 10.1002/jbm.a.37379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 02/21/2022] [Accepted: 02/26/2022] [Indexed: 12/14/2022]
Abstract
Articular cartilage degeneration is still an unsolved issue owing to its weak repairing capabilities, which usually result in fibrocartilage tissue formation. This fibrous tissue lacks of structural and bio-mechanical properties, degrading over time. Currently, arthroscopic techniques and autologous transplantation are the most used clinical procedures. However, rather than restoring cartilage integrity, these methods only postpone further cartilage deterioration. Therefore, tissue engineering strategies aimed at selecting scaffolds that remarkably support the chondrogenic differentiation of human mesenchymal stem cells (hMSCs) could represent a promising solution, but they are still challenging for researchers. In this study, the influence of two different genipin (Gp) crosslinking routes on collagen (Coll)-based scaffolds in terms of hMSCs chondrogenic differentiation and biomechanical performances was investigated. Three-dimensional (3D) porous Coll scaffolds were fabricated by freeze-drying techniques and were crosslinked with Gp following a "two-step" and an in "bulk" procedure, in order to increase the physico-mechanical stability of the structure. Chondrogenic differentiation efficacy of hMSCs and biomechanical behavior under compression forces through unconfined stress-strain tests were assessed. Coll/Gp scaffolds revealed an isotropic and highly homogeneous pore distribution along with an increase in the stiffness, also supported by the increase in the Coll denaturation temperature (Td = 57-63°C) and a significant amount of Coll and GAG deposition during the 3 weeks of chondrogenic culture. In particular, the presence of Gp in "bulk" led to a more uniform and homogenous chondral-like matrix deposition by hMSCs if compared to the results obtained from the Gp "two-step" functionalization procedure.
Collapse
Affiliation(s)
- Stefania Scialla
- Department of Engineering for Innovation, University of Salento, Lecce, Italy.,Institute of Polymers, Composites and Biomaterials - National Research Council, Naples, Italy
| | - Fabiana Gullotta
- Department of Engineering for Innovation, University of Salento, Lecce, Italy.,Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Daniela Izzo
- Department of Engineering for Innovation, University of Salento, Lecce, Italy
| | - Barbara Palazzo
- Department of Engineering for Innovation, University of Salento, Lecce, Italy.,ENEA, Division for Sustainable Materials - Research Centre of Brindisi, Brindisi, Italy
| | - Francesca Scalera
- Department of Engineering for Innovation, University of Salento, Lecce, Italy.,CNR Nanotec - Institute of Nanotechnology, Lecce, Italy
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Alessandro Sannino
- Department of Engineering for Innovation, University of Salento, Lecce, Italy
| | - Francesca Gervaso
- Department of Engineering for Innovation, University of Salento, Lecce, Italy.,CNR Nanotec - Institute of Nanotechnology, Lecce, Italy
| |
Collapse
|
152
|
Ayariga JA, Huang H, Dean D. Decellularized Avian Cartilage, a Promising Alternative for Human Cartilage Tissue Regeneration. MATERIALS (BASEL, SWITZERLAND) 2022; 15:1974. [PMID: 35269204 PMCID: PMC8911734 DOI: 10.3390/ma15051974] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/17/2022] [Accepted: 03/02/2022] [Indexed: 02/05/2023]
Abstract
Articular cartilage defects, and subsequent degeneration, are prevalent and account for the poor quality of life of most elderly persons; they are also one of the main predisposing factors to osteoarthritis. Articular cartilage is an avascular tissue and, thus, has limited capacity for healing and self-repair. Damage to the articular cartilage by trauma or pathological causes is irreversible. Many approaches to repair cartilage have been attempted with some potential; however, there is no consensus on any ideal therapy. Tissue engineering holds promise as an approach to regenerate damaged cartilage. Since cell adhesion is a critical step in tissue engineering, providing a 3D microenvironment that recapitulates the cartilage tissue is vital to inducing cartilage regeneration. Decellularized materials have emerged as promising scaffolds for tissue engineering, since this procedure produces scaffolds from native tissues that possess structural and chemical natures that are mimetic of the extracellular matrix (ECM) of the native tissue. In this work, we present, for the first time, a study of decellularized scaffolds, produced from avian articular cartilage (extracted from Gallus Gallus domesticus), reseeded with human chondrocytes, and we demonstrate for the first time that human chondrocytes survived, proliferated and interacted with the scaffolds. Morphological studies of the decellularized scaffolds revealed an interconnected, porous architecture, ideal for cell growth. Mechanical characterization showed that the decellularized scaffolds registered stiffness comparable to the native cartilage tissues. Cell growth inhibition and immunocytochemical analyses showed that the decellularized scaffolds are suitable for cartilage regeneration.
Collapse
Affiliation(s)
| | | | - Derrick Dean
- The Biomedical Engineering Program, College of Science, Technology, Engineering and Mathematics (C-STEM), Alabama State University, 1627 Hall Street, Montgomery, AL 36104, USA; (J.A.A.); (H.H.)
| |
Collapse
|
153
|
Liao HJ, Chang CH, Huang CYF, Chen HT. Potential of Using Infrapatellar–Fat–Pad–Derived Mesenchymal Stem Cells for Therapy in Degenerative Arthritis: Chondrogenesis, Exosomes, and Transcription Regulation. Biomolecules 2022; 12:biom12030386. [PMID: 35327578 PMCID: PMC8945217 DOI: 10.3390/biom12030386] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 02/04/2023] Open
Abstract
Infrapatellar fat pad–derived mesenchymal stem cells (IPFP-MSCs) are a type of adipose-derived stem cell (ADSC). They potentially contribute to cartilage regeneration and modulation of the immune microenvironment in patients with osteoarthritis (OA). The ability of IPFP-MSCs to increase chondrogenic capacity has been reported to be greater, less age dependent, and less affected by inflammatory changes than that of other MSCs. Transcription-regulatory factors strictly regulate the cartilage differentiation of MSCs. However, few studies have explored the effect of transcriptional factors on IPFP-MSC-based neocartilage formation, cartilage engineering, and tissue functionality during and after chondrogenesis. Instead of intact MSCs, MSC-derived extracellular vesicles could be used for the treatment of OA. Furthermore, exosomes are increasingly being considered the principal therapeutic agent in MSC secretions that is responsible for the regenerative and immunomodulatory functions of MSCs in cartilage repair. The present study provides an overview of advancements in enhancement strategies for IPFP-MSC chondrogenic differentiation, including the effects of transcriptional factors, the modulation of released exosomes, delivery mechanisms for MSCs, and ethical and regulatory points concerning the development of MSC products. This review will contribute to the understanding of the IPFP-MSC chondrogenic differentiation process and enable the improvement of IPFP-MSC-based cartilage tissue engineering.
Collapse
Affiliation(s)
- Hsiu-Jung Liao
- Department of Orthopedic Surgery, Far Eastern Memorial Hospital, New Taipei City 220216, Taiwan;
| | - Chih-Hung Chang
- Department of Orthopedic Surgery, Far Eastern Memorial Hospital, New Taipei City 220216, Taiwan;
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan City 320315, Taiwan
- Correspondence: (C.-H.C.); (H.-T.C.)
| | - Chi-Ying F. Huang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Hui-Ting Chen
- Department of Pharmacy, School of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Fragrance and Cosmetic Science, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Correspondence: (C.-H.C.); (H.-T.C.)
| |
Collapse
|
154
|
Sakai H, Sawada Y, Tokunaga N, Tanaka K, Nakagawa S, Sakakibara I, Ono Y, Fukada SI, Ohkawa Y, Kikugawa T, Saika T, Imai Y. Uhrf1 governs the proliferation and differentiation of muscle satellite cells. iScience 2022; 25:103928. [PMID: 35243267 PMCID: PMC8886052 DOI: 10.1016/j.isci.2022.103928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 12/06/2021] [Accepted: 02/10/2022] [Indexed: 11/19/2022] Open
Abstract
DNA methylation is an essential form of epigenetic regulation responsible for cellular identity. In muscle stem cells, termed satellite cells, DNA methylation patterns are tightly regulated during differentiation. However, it is unclear how these DNA methylation patterns affect the function of satellite cells. We demonstrate that a key epigenetic regulator, ubiquitin like with PHD and RING finger domains 1 (Uhrf1), is activated in proliferating myogenic cells but not expressed in quiescent satellite cells or differentiated myogenic cells in mice. Ablation of Uhrf1 in mouse satellite cells impairs their proliferation and differentiation, leading to failed muscle regeneration. Uhrf1-deficient myogenic cells exhibited aberrant upregulation of transcripts, including Sox9, with the reduction of DNA methylation level of their promoter and enhancer region. These findings show that Uhrf1 is a critical epigenetic regulator of proliferation and differentiation in satellite cells, by controlling cell-type-specific gene expression via maintenance of DNA methylation. Uhrf1 is activated in proliferating myogenic cells Uhrf1 in satellite cells is required for muscle regeneration Ablation of Uhrf1 in satellite cells impairs their proliferation and differentiation Uhrf1 controls cell-type-specific transcripts via maintenance of DNA methylation
Collapse
Affiliation(s)
- Hiroshi Sakai
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Toon, Ehime 791-0295, Japan
- Department of Pathophysiology, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
- Corresponding author
| | - Yuichiro Sawada
- Department of Urology, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Naohito Tokunaga
- Division of Analytical Bio-Medicine, Advanced Research Support Center, Ehime University, Toon, Ehime 791-0295, Japan
| | - Kaori Tanaka
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Fukuoka 812-0054, Japan
| | - So Nakagawa
- Department of Molecular Life Science, Tokai University School of Medicine, Isehara, Kanagawa 259-1193, Japan
| | - Iori Sakakibara
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yusuke Ono
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Honjo, Kumamoto 860-0811, Japan
| | - So-ichiro Fukada
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Fukuoka 812-0054, Japan
| | - Tadahiko Kikugawa
- Department of Urology, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Takashi Saika
- Department of Urology, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Yuuki Imai
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Toon, Ehime 791-0295, Japan
- Department of Pathophysiology, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
- Corresponding author
| |
Collapse
|
155
|
Jo S, Lee JS, Nam B, Lee YL, Kim H, Lee EY, Park YS, Kim TH. SOX9 + enthesis cells are associated with spinal ankylosis in ankylosing spondylitis. Osteoarthritis Cartilage 2022; 30:280-290. [PMID: 34826571 DOI: 10.1016/j.joca.2021.11.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 11/09/2021] [Accepted: 11/17/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Although cartilage degeneration and invasion of the subchondral bone plate in entheseal lesion has been considered to consequently lead bony ankylosis in ankylosing spondylitis (AS), no evident mechanisms are known. DESIGN To identify histopathological and physiological changes in enthesitis-related ankylosis in AS, we performed molecular characterization of transcription factors and surface markers, and transcriptome analysis with human tissues. Entheseal tissue containing subchondral bone was obtained from the facet joints of 9 patients with AS and 10 disease controls, and assessed by using differential staining techniques. Enthesis cells were isolated, characterized, stimulated with TNF and/or IL-17A, and analysed by cell-based experimental tools. RESULTS We found diffusely distributed granular tissue and cartilage in the subchondral bone in AS. Co-expression of SOX9, a specific transcription factor in cartilage, and matrix metalloproteinase 13 (MMP13) was found in the granular tissues within the subchondral bone from AS patients. Intriguingly, SOX9 expression was significantly higher in AS enthesis cells than controls and correlated with TNFR1 and IL-17RA expressions, which is important for high reactivity to TNF and IL-17A cytokines. Co-stimulation by TNF and IL-17A resulted in accelerated mineralization/calcification features, and increased OCN expression in AS enthesis cells. Furthermore, SOX9 overexpression in enthesis leads to promoting mineralization feature by TNF and IL-17A stimuli. Finally, OCN expression is elevated in the destructive enthesis of advanced AS. CONCLUSION These findings provide insight into the links between inflammation and the mineralization of entheseal tissue as the initiation of spinal ankylosis, emphasizing the importance of SOX9+ enthesis cells.
Collapse
Affiliation(s)
- S Jo
- Hanyang University Institute for Rheumatology Research, Hanyang University Hospital for Rheumatic Diseases, Seoul 04763, Republic of Korea
| | - J S Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea; GENOME INSIGHT Inc., Daejeon 34141, Republic of Korea
| | - B Nam
- Hanyang University Institute for Rheumatology Research, Hanyang University Hospital for Rheumatic Diseases, Seoul 04763, Republic of Korea
| | - Y L Lee
- Hanyang University Institute for Rheumatology Research, Hanyang University Hospital for Rheumatic Diseases, Seoul 04763, Republic of Korea
| | - H Kim
- Department of Pathology, Hanyang University Seoul Hospital, Seoul 04763, Republic of Korea
| | - E Y Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Y-S Park
- Department of Orthopedic Surgery, Guri Hospital, Hanyang University College of Medicine, Guri 11923, Republic of Korea
| | - T-H Kim
- Hanyang University Institute for Rheumatology Research, Hanyang University Hospital for Rheumatic Diseases, Seoul 04763, Republic of Korea; Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul 04763, Republic of Korea.
| |
Collapse
|
156
|
Live imaging approach of dynamic multicellular responses in ERK signaling during vertebrate tissue development. Biochem J 2022; 479:129-143. [PMID: 35050327 PMCID: PMC8883488 DOI: 10.1042/bcj20210557] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/28/2021] [Accepted: 01/05/2022] [Indexed: 11/17/2022]
Abstract
The chemical and mechanical responses of cells via the exchange of information during growth and development result in the formation of biological tissues. Information processing within the cells through the signaling pathways and networks inherent to the constituent cells has been well-studied. However, the cell signaling mechanisms responsible for generating dynamic multicellular responses in developing tissues remain unclear. Here, I review the dynamic multicellular response systems during the development and growth of vertebrate tissues based on the extracellular signal-regulated kinase (ERK) pathway. First, an overview of the function of the ERK signaling network in cells is provided, followed by descriptions of biosensors essential for live imaging of the quantification of ERK activity in tissues. Then adducing four examples, I highlight the contribution of live imaging techniques for studying the involvement of spatio-temporal patterns of ERK activity change in tissue development and growth. In addition, theoretical implications of ERK signaling are also discussed from the viewpoint of dynamic systems. This review might help in understanding ERK-mediated dynamic multicellular responses and tissue morphogenesis.
Collapse
|
157
|
Sermeus Y, Vangheel J, Geris L, Smeets B, Tylzanowski P. Mechanical Regulation of Limb Bud Formation. Cells 2022; 11:420. [PMID: 35159230 PMCID: PMC8834596 DOI: 10.3390/cells11030420] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/20/2022] [Accepted: 01/23/2022] [Indexed: 12/28/2022] Open
Abstract
Early limb bud development has been of considerable interest for the study of embryological development and especially morphogenesis. The focus has long been on biochemical signalling and less on cell biomechanics and mechanobiology. However, their importance cannot be understated since tissue shape changes are ultimately controlled by active forces and bulk tissue rheological properties that in turn depend on cell-cell interactions as well as extracellular matrix composition. Moreover, the feedback between gene regulation and the biomechanical environment is still poorly understood. In recent years, novel experimental techniques and computational models have reinvigorated research on this biomechanical and mechanobiological side of embryological development. In this review, we consider three stages of early limb development, namely: outgrowth, elongation, and condensation. For each of these stages, we summarize basic biological regulation and examine the role of cellular and tissue mechanics in the morphogenetic process.
Collapse
Affiliation(s)
- Yvenn Sermeus
- MeBioS, KU Leuven, 3000 Leuven, Belgium; (Y.S.); (J.V.); (B.S.)
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium;
| | - Jef Vangheel
- MeBioS, KU Leuven, 3000 Leuven, Belgium; (Y.S.); (J.V.); (B.S.)
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium;
| | - Liesbet Geris
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium;
- GIGA In Silico Medicine, Université de Liège, 4000 Liège, Belgium
- SBE, Department of Development and Regeneration, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Bart Smeets
- MeBioS, KU Leuven, 3000 Leuven, Belgium; (Y.S.); (J.V.); (B.S.)
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium;
| | - Przemko Tylzanowski
- SBE, Department of Development and Regeneration, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
- Laboratory of Molecular Genetics, Department of Biomedical Sciences, Medical University of Lublin, Chodzki 1, 20-093 Lublin, Poland
| |
Collapse
|
158
|
Smeeton J, Natarajan N, Anderson T, Tseng KC, Fabian P, Crump JG. Regeneration of Jaw Joint Cartilage in Adult Zebrafish. Front Cell Dev Biol 2022; 9:777787. [PMID: 35127702 PMCID: PMC8811260 DOI: 10.3389/fcell.2021.777787] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/07/2021] [Indexed: 11/17/2022] Open
Abstract
The poor intrinsic repair capacity of mammalian joint cartilage likely contributes to the high incidence of arthritis worldwide. Adult zebrafish can regenerate many structures that show limited or no healing capacity in mammals, including the jawbone. To test whether zebrafish can also regenerate damaged joints, we developed a surgical injury model in which the zebrafish jaw joint is destabilized via transection of the major jaw joint ligament, the interopercular-mandibular (IOM). Unilateral transection of the IOM ligament in 1-year-old fish resulted in an initial reduction of jaw joint cartilage by 14 days, with full regeneration of joint cartilage by 28 days. Joint cartilage regeneration involves the re-entry of articular chondrocytes into the cell cycle and the upregulated expression of sox10, a marker of developing chondrocytes in the embryo that becomes restricted to a subset of joint chondrocytes in adults. Genetic ablation of these sox10-expressing chondrocytes shows that they are essential for joint cartilage regeneration. To uncover the potential source of new chondrocytes during joint regeneration, we performed single-cell RNA sequencing of the uninjured adult jaw joint and identified multiple skeletal, connective tissue, and fibroblast subtypes. In particular, we uncovered a joint-specific periosteal population expressing coch and grem1a, with the jaw joint chondrocytes marked by grem1a expression during regeneration. Our findings demonstrate the capacity of zebrafish to regenerate adult joint cartilage and identify candidate cell types that can be tested for their roles in regenerative response.
Collapse
Affiliation(s)
- Joanna Smeeton
- Department of Rehabilitation and Regenerative Medicine, Columbia Stem Cell Initiative, Columbia University Irving Medical Center, Columbia University, New York, NY, United States
- Department of Genetics and Development, Columbia Stem Cell Initiative, Columbia University Irving Medical Center, Columbia University, New York, NY, United States
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Natasha Natarajan
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Troy Anderson
- Department of Rehabilitation and Regenerative Medicine, Columbia Stem Cell Initiative, Columbia University Irving Medical Center, Columbia University, New York, NY, United States
- Department of Genetics and Development, Columbia Stem Cell Initiative, Columbia University Irving Medical Center, Columbia University, New York, NY, United States
| | - Kuo-Chang Tseng
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Peter Fabian
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - J. Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
159
|
Nonsuwan P, Nishijima N, Sakakibara K, Nakaji-Hirabayashi T, Yoshikawa C. Concentrated polymer brush-modified cellulose nanofibers promote chondrogenic differentiation of human mesenchymal stem cells by controlling self-assembly. J Mater Chem B 2022; 10:2444-2453. [PMID: 35045146 DOI: 10.1039/d1tb02307a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In order to develop new three-dimensional (3D) cell culture systems for articular cartilage regeneration, concentrated poly(styrene sulfonate sodium salt) brush-modified cellulose nanofibers were employed as building blocks for the self-assembly of human mesenchymal stem cells (hMSCs). Unique 3D cellular structures, such as giant spheres and sheets, were formed by controlling hMSC self-assembly.
Collapse
Affiliation(s)
- Punnida Nonsuwan
- Research Center for Functional Materials, National Institute for Materials Science (NIMS), Tsukuba, Ibaraki 305-0047, Japan.
| | - Nanami Nishijima
- Research Center for Functional Materials, National Institute for Materials Science (NIMS), Tsukuba, Ibaraki 305-0047, Japan. .,Faculty of Engineering, Academic Assembly, University of Toyama, 3190 Gofuku, Toyama, Toyama 930-8555, Japan
| | - Keita Sakakibara
- Research Institute for Sustainable Chemistry, National Institute of Advanced Industrial Science and Technology (AIST), 3-11-32 Kagamiyama, Higashi-hiroshima, Hiroshima 739-0046, Japan
| | - Tadashi Nakaji-Hirabayashi
- Research Center for Functional Materials, National Institute for Materials Science (NIMS), Tsukuba, Ibaraki 305-0047, Japan. .,Faculty of Engineering, Academic Assembly, University of Toyama, 3190 Gofuku, Toyama, Toyama 930-8555, Japan.,Graduate School of Innovative Life Science, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan
| | - Chiaki Yoshikawa
- Research Center for Functional Materials, National Institute for Materials Science (NIMS), Tsukuba, Ibaraki 305-0047, Japan.
| |
Collapse
|
160
|
Liu NQ, Lin Y, Li L, Lu J, Geng D, Zhang J, Jashashvili T, Buser Z, Magallanes J, Tassey J, Shkhyan R, Sarkar A, Lopez N, Lee S, Lee Y, Wang L, Petrigliano FA, Van Handel B, Lyons K, Evseenko D. gp130/STAT3 signaling is required for homeostatic proliferation and anabolism in postnatal growth plate and articular chondrocytes. Commun Biol 2022; 5:64. [PMID: 35039652 PMCID: PMC8763901 DOI: 10.1038/s42003-021-02944-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/03/2021] [Indexed: 02/05/2023] Open
Abstract
Growth of long bones and vertebrae is maintained postnatally by a long-lasting pool of progenitor cells. Little is known about the molecular mechanisms that regulate the output and maintenance of the cells that give rise to mature cartilage. Here we demonstrate that postnatal chondrocyte-specific deletion of a transcription factor Stat3 results in severely reduced proliferation coupled with increased hypertrophy, growth plate fusion, stunting and signs of progressive dysfunction of the articular cartilage. This effect is dimorphic, with females more strongly affected than males. Chondrocyte-specific deletion of the IL-6 family cytokine receptor gp130, which activates Stat3, phenocopied Stat3-deletion; deletion of Lifr, one of many co-receptors that signals through gp130, resulted in a milder phenotype. These data define a molecular circuit that regulates chondrogenic cell maintenance and output and reveals a pivotal positive function of IL-6 family cytokines in the skeletal system with direct implications for skeletal development and regeneration.
Collapse
Affiliation(s)
- Nancy Q. Liu
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA
| | - Yucheng Lin
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA ,grid.89957.3a0000 0000 9255 8984Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006 China ,grid.263826.b0000 0004 1761 0489Department of Orthopaedic Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009 China
| | - Liangliang Li
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA ,grid.89957.3a0000 0000 9255 8984Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006 China ,grid.89957.3a0000 0000 9255 8984Department of Orthopedics, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu 211100 China
| | - Jinxiu Lu
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA
| | - Dawei Geng
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA ,grid.89957.3a0000 0000 9255 8984Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006 China ,grid.89957.3a0000 0000 9255 8984Department of Orthopaedic Surgery, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu 211166 China
| | - Jiankang Zhang
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA ,grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 China
| | - Tea Jashashvili
- grid.42505.360000 0001 2156 6853Department of Radiology, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA
| | - Zorica Buser
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA
| | - Jenny Magallanes
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA
| | - Jade Tassey
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA
| | - Ruzanna Shkhyan
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA
| | - Arijita Sarkar
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA
| | - Noah Lopez
- grid.19006.3e0000 0000 9632 6718Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angles (UCLA), Los Angeles, CA USA
| | - Siyoung Lee
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA
| | - Youngjoo Lee
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA
| | - Liming Wang
- grid.89957.3a0000 0000 9255 8984Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006 China ,grid.89957.3a0000 0000 9255 8984Institute of Digital Medicine, Nanjing Medical University, Nanjing, Jiangsu 210006 China
| | - Frank A. Petrigliano
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA ,grid.42505.360000 0001 2156 6853Department of Stem Cell Research and Regenerative Medicine, USC, Los Angeles, CA 90033 USA
| | - Ben Van Handel
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA
| | - Karen Lyons
- grid.19006.3e0000 0000 9632 6718Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angles (UCLA), Los Angeles, CA USA
| | - Denis Evseenko
- grid.42505.360000 0001 2156 6853Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033 USA ,grid.42505.360000 0001 2156 6853Department of Stem Cell Research and Regenerative Medicine, USC, Los Angeles, CA 90033 USA
| |
Collapse
|
161
|
Yu L, Lin YL, Yan M, Li T, Wu EY, Zimmel K, Qureshi O, Falck A, Sherman KM, Huggins SS, Hurtado DO, Suva LJ, Gaddy D, Cai J, Brunauer R, Dawson LA, Muneoka K. Hyaline cartilage differentiation of fibroblasts in regeneration and regenerative medicine. Development 2022; 149:274141. [PMID: 35005773 PMCID: PMC8917415 DOI: 10.1242/dev.200249] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/15/2021] [Indexed: 11/30/2022]
Abstract
Amputation injuries in mammals are typically non-regenerative; however, joint regeneration is stimulated by BMP9 treatment, indicating the presence of latent articular chondrocyte progenitor cells. BMP9 induces a battery of chondrogenic genes in vivo, and a similar response is observed in cultures of amputation wound cells. Extended cultures of BMP9-treated cells results in differentiation of hyaline cartilage, and single cell RNAseq analysis identified wound fibroblasts as BMP9 responsive. This culture model was used to identify a BMP9-responsive adult fibroblast cell line and a culture strategy was developed to engineer hyaline cartilage for engraftment into an acutely damaged joint. Transplanted hyaline cartilage survived engraftment and maintained a hyaline cartilage phenotype, but did not form mature articular cartilage. In addition, individual hypertrophic chondrocytes were identified in some samples, indicating that the acute joint injury site can promote osteogenic progression of engrafted hyaline cartilage. The findings identify fibroblasts as a cell source for engineering articular cartilage and establish a novel experimental strategy that bridges the gap between regeneration biology and regenerative medicine. Summary:In vivo articular cartilage regeneration serves as a model to develop novel approaches for engineering cartilage to repair damaged joints and identifies fibroblasts as a BMP9-inducible chondroprogenitor.
Collapse
Affiliation(s)
- Ling Yu
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Yu-Lieh Lin
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Mingquan Yan
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Tao Li
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, People's Republic of China
| | - Emily Y. Wu
- Dewpoint Therapeutics, 6 Tide Street, Suite 300, Boston, MA 02210, USA
| | - Katherine Zimmel
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Osama Qureshi
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Alyssa Falck
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Kirby M. Sherman
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Shannon S. Huggins
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Daniel Osorio Hurtado
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Larry J. Suva
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Dana Gaddy
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - James Cai
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Regina Brunauer
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Lindsay A. Dawson
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Ken Muneoka
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
162
|
Chen H, Cui Y, Zhang D, Xie J, Zhou X. The role of fibroblast growth factor 8 in cartilage development and disease. J Cell Mol Med 2022; 26:990-999. [PMID: 35001536 PMCID: PMC8831980 DOI: 10.1111/jcmm.17174] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 02/05/2023] Open
Abstract
Fibroblast growth factor 8 (FGF‐8), also known as androgen‐induced growth factor (AIGF), is presumed to be a potent mitogenic cytokine that plays important roles in early embryonic development, brain formation and limb development. In the bone environment, FGF‐8 produced or received by chondrocyte precursor cells binds to fibroblast growth factor receptor (FGFR), causing different levels of activation of downstream signalling pathways, such as phospholipase C gamma (PLCγ)/Ca2+, RAS/mitogen‐activated protein kinase‐extracellular regulated protein kinases (RAS/MAPK‐MEK‐ERK), and Wnt‐β‐catenin‐Axin2 signalling, and ultimately controlling chondrocyte proliferation, differentiation, cell survival and migration. However, the molecular mechanism of FGF‐8 in normal or pathological cartilage remains unclear, and thus, FGF‐8 represents a novel exploratory target for studies of chondrocyte development and cartilage disease progression. In this review, studies assessing the relationship between FGF‐8 and chondrocytes that have been published in the past 5 years are systematically summarized to determine the probable mechanism and physiological effect of FGF‐8 on chondrocytes. Based on the existing research results, a therapeutic regimen targeting FGF‐8 is proposed to explore the possibility of treating chondrocyte‐related diseases.
Collapse
Affiliation(s)
- Haoran Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yujia Cui
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
163
|
Chabronova A, van den Akker GGH, Meekels-Steinbusch MMF, Friedrich F, Cremers A, Surtel DAM, Peffers MJ, van Rhijn LW, Lausch E, Zabel B, Caron MMJ, Welting TJM. Uncovering pathways regulating chondrogenic differentiation of CHH fibroblasts. Noncoding RNA Res 2022; 6:211-224. [PMID: 34988338 PMCID: PMC8688813 DOI: 10.1016/j.ncrna.2021.12.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/08/2021] [Accepted: 12/08/2021] [Indexed: 02/08/2023] Open
Abstract
Mutations in the non-coding snoRNA component of mitochondrial RNA processing endoribonuclease (RMRP) are the cause of cartilage-hair hypoplasia (CHH). CHH is a rare form of metaphyseal chondrodysplasia characterized by disproportionate short stature and abnormal growth plate development. The process of chondrogenic differentiation within growth plates of long bones is vital for longitudinal bone growth. However, molecular mechanisms behind impaired skeletal development in CHH patients remain unclear. We employed a transdifferentiation model (FDC) combined with whole transcriptome analysis to investigate the chondrogenic transdifferentiation capacity of CHH fibroblasts and to examine pathway regulation in CHH cells during chondrogenic differentiation. We established that the FDC transdifferentiation model is a relevant in vitro model of chondrogenic differentiation, with an emphasis on the terminal differentiation phase, which is crucial for longitudinal bone growth. We demonstrated that CHH fibroblasts are capable of transdifferentiating into chondrocyte-like cells, and show a reduced commitment to terminal differentiation. We also found a number of key factors of BMP, FGF, and IGF-1 signalling axes to be significantly upregulated in CHH cells during the chondrogenic transdifferentiation. Our results support postulated conclusions that RMRP has pleiotropic functions and profoundly affects multiple aspects of cell fate and signalling. Our findings shed light on the consequences of pathological CHH mutations in snoRNA RMRP during chondrogenic differentiation and the relevance and roles of non-coding RNAs in genetic diseases in general.
Collapse
Affiliation(s)
- Alzbeta Chabronova
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, MUMC+, 6202, AZ, Maastricht, the Netherlands
| | - Guus G H van den Akker
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, MUMC+, 6202, AZ, Maastricht, the Netherlands
| | - Mandy M F Meekels-Steinbusch
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, MUMC+, 6202, AZ, Maastricht, the Netherlands
| | - Franziska Friedrich
- Department of Pediatrics, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andy Cremers
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, MUMC+, 6202, AZ, Maastricht, the Netherlands
| | - Don A M Surtel
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, MUMC+, 6202, AZ, Maastricht, the Netherlands
| | - Mandy J Peffers
- Institute of Life Course and Medical Sciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Lodewijk W van Rhijn
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, MUMC+, 6202, AZ, Maastricht, the Netherlands
| | - Ekkehart Lausch
- Department of Pediatrics, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bernhard Zabel
- Medical Faculty, Otto van Guericke University of Magdeburg, 39106, Magdeburg, Germany
| | - Marjolein M J Caron
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, MUMC+, 6202, AZ, Maastricht, the Netherlands
| | - Tim J M Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, MUMC+, 6202, AZ, Maastricht, the Netherlands
| |
Collapse
|
164
|
Bauza‐Mayol G, Quintela M, Brozovich A, Hopson M, Shaikh S, Cabrera F, Shi A, Niclot FB, Paradiso F, Combellack E, Jovic T, Rees P, Tasciotti E, Francis LW, Mcculloch P, Taraballi F. Biomimetic Scaffolds Modulate the Posttraumatic Inflammatory Response in Articular Cartilage Contributing to Enhanced Neoformation of Cartilaginous Tissue In Vivo. Adv Healthc Mater 2022; 11:e2101127. [PMID: 34662505 PMCID: PMC11469755 DOI: 10.1002/adhm.202101127] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 09/28/2021] [Indexed: 12/13/2022]
Abstract
Focal chondral lesions of the knee are the most frequent type of trauma in younger patients and are associated with a high risk of developing early posttraumatic osteoarthritis. The only current clinical solutions include microfracture, osteochondral grafting, and autologous chondrocyte implantation. Cartilage tissue engineering based on biomimetic scaffolds has become an appealing strategy to repair cartilage defects. Here, a chondrogenic collagen-chondroitin sulfate scaffold is tested in an orthotopic Lapine in vivo model to understand the beneficial effects of the immunomodulatory biomaterial on the full chondral defect. Using a combination of noninvasive imaging techniques, histological and whole transcriptome analysis, the scaffolds are shown to enhance the formation of cartilaginous tissue and suppression of host cartilage degeneration, while also supporting tissue integration and increased tissue regeneration over a 12 weeks recovery period. The results presented suggest that biomimetic materials could be a clinical solution for cartilage tissue repair, due to their ability to modulate the immune environment in favor of regenerative processes and suppression of cartilage degeneration.
Collapse
Affiliation(s)
- Guillermo Bauza‐Mayol
- Center for Musculoskeletal RegenerationHouston Methodist Research Institute6670 Bertner Ave.HoustonTX77030USA
- Orthopedics & Sports MedicineHouston Methodist Hospital6550 Fannin St.HoustonTX77030USA
- Reproductive Biology and Gynaecological Oncology GroupSwansea University Medical SchoolSingleton ParkSwanseaSA2 8PPUK
| | - Marcos Quintela
- Reproductive Biology and Gynaecological Oncology GroupSwansea University Medical SchoolSingleton ParkSwanseaSA2 8PPUK
| | - Ava Brozovich
- Center for Musculoskeletal RegenerationHouston Methodist Research Institute6670 Bertner Ave.HoustonTX77030USA
- Orthopedics & Sports MedicineHouston Methodist Hospital6550 Fannin St.HoustonTX77030USA
- Texas A&M College of MedicineBryanTX77807USA
| | - Michael Hopson
- Orthopedics & Sports MedicineHouston Methodist Hospital6550 Fannin St.HoustonTX77030USA
| | - Shazad Shaikh
- Orthopedics & Sports MedicineHouston Methodist Hospital6550 Fannin St.HoustonTX77030USA
| | - Fernando Cabrera
- Center for Musculoskeletal RegenerationHouston Methodist Research Institute6670 Bertner Ave.HoustonTX77030USA
- Orthopedics & Sports MedicineHouston Methodist Hospital6550 Fannin St.HoustonTX77030USA
| | - Aaron Shi
- Center for Musculoskeletal RegenerationHouston Methodist Research Institute6670 Bertner Ave.HoustonTX77030USA
- Orthopedics & Sports MedicineHouston Methodist Hospital6550 Fannin St.HoustonTX77030USA
| | - Federica Banche Niclot
- Center for Musculoskeletal RegenerationHouston Methodist Research Institute6670 Bertner Ave.HoustonTX77030USA
- Polytechnic of TurinDepartment of Applied Science and TechnologyCorso Duca degli Abruzzi 24Torino10129Italy
| | - Francesca Paradiso
- Center for Musculoskeletal RegenerationHouston Methodist Research Institute6670 Bertner Ave.HoustonTX77030USA
- Orthopedics & Sports MedicineHouston Methodist Hospital6550 Fannin St.HoustonTX77030USA
- Reproductive Biology and Gynaecological Oncology GroupSwansea University Medical SchoolSingleton ParkSwanseaSA2 8PPUK
| | - Emman Combellack
- Reconstructive Surgery and Regenerative Medicine Research GroupSwansea University Medical SchoolSingleton ParkSwanseaSA2 8PPUK
| | - Tom Jovic
- Reconstructive Surgery and Regenerative Medicine Research GroupSwansea University Medical SchoolSingleton ParkSwanseaSA2 8PPUK
| | - Paul Rees
- Orthopedics & Sports MedicineHouston Methodist Hospital6550 Fannin St.HoustonTX77030USA
| | | | - Lewis W. Francis
- Center for Musculoskeletal RegenerationHouston Methodist Research Institute6670 Bertner Ave.HoustonTX77030USA
| | - Patrick Mcculloch
- Orthopedics & Sports MedicineHouston Methodist Hospital6550 Fannin St.HoustonTX77030USA
| | - Francesca Taraballi
- Center for Musculoskeletal RegenerationHouston Methodist Research Institute6670 Bertner Ave.HoustonTX77030USA
- Orthopedics & Sports MedicineHouston Methodist Hospital6550 Fannin St.HoustonTX77030USA
| |
Collapse
|
165
|
Kumar N, Saraber P, Ding Z, Kusumbe AP. Diversity of Vascular Niches in Bones and Joints During Homeostasis, Ageing, and Diseases. Front Immunol 2021; 12:798211. [PMID: 34975909 PMCID: PMC8718446 DOI: 10.3389/fimmu.2021.798211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/19/2021] [Indexed: 12/29/2022] Open
Abstract
The bones and joints in the skeletal system are composed of diverse cell types, including vascular niches, bone cells, connective tissue cells and mineral deposits and regulate whole-body homeostasis. The capacity of maintaining strength and generation of blood lineages lies within the skeletal system. Bone harbours blood and immune cells and their progenitors, and vascular cells provide several immune cell type niches. Blood vessels in bone are phenotypically and functionally diverse, with distinct capillary subtypes exhibiting striking changes with age. The bone vasculature has a special impact on osteogenesis and haematopoiesis, and dysregulation of the vasculature is associated with diverse blood and bone diseases. Ageing is associated with perturbed haematopoiesis, loss of osteogenesis, increased adipogenesis and diminished immune response and immune cell production. Endothelial and perivascular cells impact immune cell production and play a crucial role during inflammation. Here, we discuss normal and maladapted vascular niches in bone during development, homeostasis, ageing and bone diseases such as rheumatoid arthritis and osteoarthritis. Further, we discuss the role of vascular niches during bone malignancy.
Collapse
Affiliation(s)
| | | | | | - Anjali P. Kusumbe
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), Tissue and Tumor Microenvironments Group, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
166
|
Ott C, Pappritz K, Hegemann N, John C, Jeuthe S, McAlpine CS, Iwamoto Y, Lauryn JH, Klages J, Klopfleisch R, Van Linthout S, Swirski F, Nahrendorf M, Kintscher U, Grune T, Kuebler WM, Grune J. Spontaneous Degenerative Aortic Valve Disease in New Zealand Obese Mice. J Am Heart Assoc 2021; 10:e023131. [PMID: 34779224 PMCID: PMC9075397 DOI: 10.1161/jaha.121.023131] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Degenerative aortic valve (AoV) disease and resulting aortic stenosis are major clinical health problems. Murine models of valve disease are rare, resulting in a translational knowledge gap on underlying mechanisms, functional consequences, and potential therapies. Naïve New Zealand obese (NZO) mice were recently found to have a dramatic decline of left ventricular (LV) function at early age. Therefore, we aimed to identify the underlying cause of reduced LV function in NZO mice. Methods and Results Cardiac function and pulmonary hemodynamics of NZO and age-matched C57BL/6J mice were monitored by serial echocardiographic examinations. AoVs in NZO mice demonstrated extensive thickening, asymmetric aortic leaflet formation, and cartilaginous transformation of the valvular stroma. Doppler echocardiography of the aorta revealed increased peak velocity profiles, holodiastolic flow reversal, and dilatation of the ascending aorta, consistent with aortic stenosis and regurgitation. Compensated LV hypertrophy deteriorated to decompensated LV failure and remodeling, as indicated by increased LV mass, interstitial fibrosis, and inflammatory cell infiltration. Elevated LV pressures in NZO mice were associated with lung congestion and cor pulmonale, evident as right ventricular dilatation, decreased right ventricular function, and increased mean right ventricular systolic pressure, indicative for the development of pulmonary hypertension and ultimately right ventricular failure. Conclusions NZO mice demonstrate as a novel murine model to spontaneously develop degenerative AoV disease, aortic stenosis, and the associated end organ damages of both ventricles and the lung. Closely mimicking the clinical scenario of degenerative AoV disease, the model may facilitate a better mechanistic understanding and testing of novel treatment strategies in degenerative AoV disease.
Collapse
Affiliation(s)
- Christiane Ott
- Department of Molecular Toxicology German Institute of Human Nutrition Potsdam-Rehbruecke Germany.,German Centre for Cardiovascular Research (partner site Berlin) Berlin Germany
| | - Kathleen Pappritz
- German Centre for Cardiovascular Research (partner site Berlin) Berlin Germany.,Berlin Institute of Health Center for Regenerative Therapies and Berlin-Brandenburg Center for Regenerative Therapies Charité-Universitätsmedizin BerlinCampus Virchow Klinikum Berlin Germany
| | - Niklas Hegemann
- German Centre for Cardiovascular Research (partner site Berlin) Berlin Germany.,Institute of Physiology Charité-Universitätsmedizin Berlin Berlin Germany
| | - Cathleen John
- Department of Molecular Toxicology German Institute of Human Nutrition Potsdam-Rehbruecke Germany.,German Centre for Cardiovascular Research (partner site Berlin) Berlin Germany
| | - Sarah Jeuthe
- German Centre for Cardiovascular Research (partner site Berlin) Berlin Germany.,Department of Medicine/Cardiology Deutsches Herzzentrum Berlin Berlin Germany.,Max-Delbrück Center for Molecular Medicine in the Helmholtz Association Berlin Germany
| | - Cameron S McAlpine
- Center for Systems Biology Massachusetts General Hospital and Harvard Medical School Boston MA
| | - Yoshiko Iwamoto
- Center for Systems Biology Massachusetts General Hospital and Harvard Medical School Boston MA
| | - Jonathan H Lauryn
- German Centre for Cardiovascular Research (partner site Berlin) Berlin Germany.,Institute of Physiology Charité-Universitätsmedizin Berlin Berlin Germany
| | - Jan Klages
- Department of Anesthesiology Deutsches Herzzentrum Berlin Berlin Germany
| | - Robert Klopfleisch
- Department of Veterinary Pathology Freie Universität Berlin Berlin Germany
| | - Sophie Van Linthout
- German Centre for Cardiovascular Research (partner site Berlin) Berlin Germany.,Berlin Institute of Health Center for Regenerative Therapies and Berlin-Brandenburg Center for Regenerative Therapies Charité-Universitätsmedizin BerlinCampus Virchow Klinikum Berlin Germany.,Department of Cardiology Charité-Universitätsmedizin BerlinCampus Virchow Klinikum Berlin Germany
| | - Fil Swirski
- Center for Systems Biology Massachusetts General Hospital and Harvard Medical School Boston MA
| | - Matthias Nahrendorf
- Center for Systems Biology Massachusetts General Hospital and Harvard Medical School Boston MA
| | - Ulrich Kintscher
- German Centre for Cardiovascular Research (partner site Berlin) Berlin Germany.,Center for Cardiovascular Research/Institute of Pharmacology Charité-Universitätsmedizin Berlin Berlin Germany
| | - Tilman Grune
- Department of Molecular Toxicology German Institute of Human Nutrition Potsdam-Rehbruecke Germany.,German Centre for Cardiovascular Research (partner site Berlin) Berlin Germany.,German Center for Diabetes Research München-Neuherberg Germany.,Institute of Nutritional Science University of Potsdam Nuthetal Germany
| | - Wolfgang M Kuebler
- German Centre for Cardiovascular Research (partner site Berlin) Berlin Germany.,Institute of Physiology Charité-Universitätsmedizin Berlin Berlin Germany.,Departments of Surgery and Physiology University of Toronto and Keenan Research Centre for Biomedical Science of St. Michael's Toronto Canada
| | - Jana Grune
- German Centre for Cardiovascular Research (partner site Berlin) Berlin Germany.,Institute of Physiology Charité-Universitätsmedizin Berlin Berlin Germany.,Center for Systems Biology Massachusetts General Hospital and Harvard Medical School Boston MA.,Center for Cardiovascular Research/Institute of Pharmacology Charité-Universitätsmedizin Berlin Berlin Germany
| |
Collapse
|
167
|
Vesela B, Svandova E, Ramesova A, Kratochvilova A, Tucker AS, Matalova E. Caspase Inhibition Affects the Expression of Autophagy-Related Molecules in Chondrocytes. Cartilage 2021; 13:956S-968S. [PMID: 32627581 PMCID: PMC8804809 DOI: 10.1177/1947603520938444] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Objective. Caspases, cysteine proteases traditionally associated with apoptosis and inflammation, have recently been identified as important regulators of autophagy and reported within the growth plate, a cartilaginous part of the developing bone. The aim of this research was to identify novel autophagy-related molecules affected by inhibition of pro-apoptotic caspases in chondrocytes. Design. Chondrocyte micromasses derived from mouse limb buds were treated with pharmacological inhibitors of caspases. Autophagy-related gene expression was examined and possible novel molecules were confirmed by real-time polymerase chain reaction and immunocytofluorescence. Individual caspases inhibitors were used to identify the effect of specific caspases. Results. Chondrogenesis accompanied by caspase activation and autophagy progression was confirmed in micromass cultures. Expression of several autophagy-associated genes was significantly altered in the caspases inhibitors treated groups with the most prominent decrease for Pik3cg and increase of Tnfsf10. The results showed the specific pro-apoptotic caspases that play a role in these effects. Importantly, use of caspase inhibitors mimicked changes triggered by an autophagy stimulator, rapamycin, linking loss of caspase activity to an increase in autophagy. Conclusion. Caspase inhibition significantly affects regulation of autophagy-related genes in chondrocytes cultures. Detected markers are of importance in diagnostics and thus the data presented here open new perspectives in the field of cartilage development and degradation.
Collapse
Affiliation(s)
- Barbora Vesela
- Department of Physiology, University of
Veterinary and Pharmaceutical Sciences, Brno, Czech Republic,Institute of Animal Physiology and
Genetics, Czech Academy of Sciences, Brno, Czech Republic,Barbora Vesela, Institute of Animal
Physiology and Genetics, Czech Academy of Sciences, v.v.i., Veveri 97, Brno
60200, Czech Republic.
| | - Eva Svandova
- Department of Physiology, University of
Veterinary and Pharmaceutical Sciences, Brno, Czech Republic,Institute of Animal Physiology and
Genetics, Czech Academy of Sciences, Brno, Czech Republic
| | - Alice Ramesova
- Department of Physiology, University of
Veterinary and Pharmaceutical Sciences, Brno, Czech Republic
| | - Adela Kratochvilova
- Institute of Animal Physiology and
Genetics, Czech Academy of Sciences, Brno, Czech Republic
| | - Abigail S. Tucker
- Centre for Craniofacial and Regenerative
Biology, King’s College London, London, UK
| | - Eva Matalova
- Department of Physiology, University of
Veterinary and Pharmaceutical Sciences, Brno, Czech Republic,Institute of Animal Physiology and
Genetics, Czech Academy of Sciences, Brno, Czech Republic
| |
Collapse
|
168
|
Abraham DM, Herman C, Witek L, Cronstein BN, Flores RL, Coelho PG. Self-assembling human skeletal organoids for disease modeling and drug testing. J Biomed Mater Res B Appl Biomater 2021; 110:871-884. [PMID: 34837719 DOI: 10.1002/jbm.b.34968] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/02/2021] [Accepted: 11/13/2021] [Indexed: 01/09/2023]
Abstract
Skeletal conditions represent a considerable challenge to health systems globally. Barriers to effective therapeutic development include a lack of accurate preclinical tissue and disease models. Most recently, work was attempted to present a novel whole organ approach to modeling human bone and cartilage tissues. These self-assembling skeletal organoids mimic the cellular milieu and extracellular organization present in native tissues. Bone organoids demonstrated osteogenesis and micro vessel formation, and cartilage organoids showed evidence of cartilage development and maturation. Skeletal organoids derived from both bone and cartilage tissues yielded spontaneous polarization of their cartilaginous and bone components. Using these hybrid skeletal organoids, we successfully generated "mini joint" cultures, which we used to model inflammatory disease and test Adenosine (A2A ) receptor agonists as a therapeutic agent. The work and respective results indicated that skeletal organoids can be an effective biological model for tissue development and disease as well as to test therapeutic agents.
Collapse
Affiliation(s)
- Diana M Abraham
- Department of Biomaterials, New York University College of Dentistry, New York, New York, USA
| | - Calvin Herman
- Department of Biomaterials, New York University College of Dentistry, New York, New York, USA
| | - Lukasz Witek
- Department of Biomaterials, New York University College of Dentistry, New York, New York, USA.,Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, New York, USA
| | - Bruce N Cronstein
- Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Roberto L Flores
- Hansjörg Wyss Department of Plastic Surgery, New York University Grossman School of Medicine, New York, New York, USA
| | - Paulo G Coelho
- Department of Biomaterials, New York University College of Dentistry, New York, New York, USA.,Hansjörg Wyss Department of Plastic Surgery, New York University Grossman School of Medicine, New York, New York, USA.,Department of Mechanical and Aerospace Engineering, New York University Tandon School of Engineering, Brooklyn, New York, USA
| |
Collapse
|
169
|
Hamada A, Mukasa H, Taguchi Y, Akagi E, Obayashi F, Yamasaki S, Kanda T, Koizumi K, Toratani S, Okamoto T. Identification of a familial cleidocranial dysplasia with a novel RUNX2 mutation and establishment of patient-derived induced pluripotent stem cells. Odontology 2021; 110:444-451. [PMID: 34779963 PMCID: PMC9170643 DOI: 10.1007/s10266-021-00674-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 10/31/2021] [Indexed: 11/24/2022]
Abstract
Cleidocranial dysplasia (CCD) is an autosomal dominant hereditary disease associated with the gene RUNX2. Disease-specific induced pluripotent stem cells (iPSCs) have emerged as a useful resource to further study human hereditary diseases such as CCD. In this study, we identified a novel CCD-specific RUNX2 mutation and established iPSCs with this mutation. Biopsies were obtained from familial CCD patients and mutation analyses were performed through Sanger sequencing and next generation sequencing. CCD-specific human iPSCs (CCD-hiPSCs) were established and maintained under completely defined serum, feeder, and integration-free condition using a non-integrating replication-defective Sendai virus vector. We identified the novel mutation RUNX2_c.371C>G and successfully established CCD-hiPSCs. The CCD-hiPSCs inherited the same mutation, possessed pluripotency, and showed the ability to differentiate the three germ layers. We concluded that RUNX2_c.371C>G was likely pathogenic because our results, derived from next generation sequencing, are supported by actual clinical evidence, familial tracing, and genetic data. Thus, we concluded that hiPSCs with a novel CCD-specific RUNX2 mutation are viable as a resource for future studies on CCD.
Collapse
Affiliation(s)
- Atsuko Hamada
- Department of Molecular Oral Medicine and Maxillofacial Surgery, Division of Applied Life Science, Graduate Institute of Biomedical and Health Science, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima-city, Hiroshima, 734-8553, Japan.
| | - Hanae Mukasa
- Department of Molecular Oral Medicine and Maxillofacial Surgery, Division of Applied Life Science, Graduate Institute of Biomedical and Health Science, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima-city, Hiroshima, 734-8553, Japan.,Mukasa Dental Clinic, Kanagawa, Japan
| | - Yuki Taguchi
- Department of Molecular Oral Medicine and Maxillofacial Surgery, Division of Applied Life Science, Graduate Institute of Biomedical and Health Science, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima-city, Hiroshima, 734-8553, Japan
| | - Eri Akagi
- Department of Molecular Oral Medicine and Maxillofacial Surgery, Division of Applied Life Science, Graduate Institute of Biomedical and Health Science, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima-city, Hiroshima, 734-8553, Japan
| | - Fumitaka Obayashi
- Department of Molecular Oral Medicine and Maxillofacial Surgery, Division of Applied Life Science, Graduate Institute of Biomedical and Health Science, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima-city, Hiroshima, 734-8553, Japan
| | - Sachiko Yamasaki
- Department of Molecular Oral Medicine and Maxillofacial Surgery, Division of Applied Life Science, Graduate Institute of Biomedical and Health Science, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima-city, Hiroshima, 734-8553, Japan
| | - Taku Kanda
- Department of Molecular Oral Medicine and Maxillofacial Surgery, Division of Applied Life Science, Graduate Institute of Biomedical and Health Science, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima-city, Hiroshima, 734-8553, Japan
| | - Koichi Koizumi
- Department of Molecular Oral Medicine and Maxillofacial Surgery, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | - Shigeaki Toratani
- Department of Molecular Oral Medicine and Maxillofacial Surgery, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | - Tetsuji Okamoto
- Department of Molecular Oral Medicine and Maxillofacial Surgery, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan.,School of Medical Sciences, The University of East Asia, Shimonoseki, Yamaguchi, 751-8503, Japan
| |
Collapse
|
170
|
Anand R, Nimi N, Sivadas VP, Merlin Rajesh Lal LP, Nair PD. Dual crosslinked pullulan-gelatin cryogel scaffold for chondrocyte-mediated cartilage repair: synthesis, characterization and in vitroevaluation. Biomed Mater 2021; 17. [PMID: 34700303 DOI: 10.1088/1748-605x/ac338b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 10/26/2021] [Indexed: 11/11/2022]
Abstract
Cryogels, a subset of hydrogels, have recently drawn attention for cartilage tissue engineering due to its inherent microporous architecture and good mechanical properties. In this study a dual crosslinked pullulan-gelatin cryogel (PDAG) scaffold was synthesized by crosslinking gelatin with oxidized pullulan by Schiff's base reaction followed by cryogelation. Chondrocytes seeded within the PDAG scaffolds and cultured for 21 din vitrodemonstrated enhanced cell proliferation, enhanced production of cartilage-specific extracellular matrix and up-regulated sulfated glycosaminoglycan without altering the articular chondrocyte phenotype. Quantitative reverse transcription-polymerase chain reaction-based gene expression studies, immunofluorescence, and histological studies demonstrated that the PDAG scaffold significantly enhanced the expression of chondrogenic marker genes such as type II collagen, aggrecan, and SOX9. Taken together, these results demonstrated that PDAG scaffold prepared by sequential Schiff's base reaction and cryogelation would be a promising cell-responsive scaffold for cartilage tissue engineering applications.
Collapse
Affiliation(s)
- Resmi Anand
- Division of Tissue Engineering and Regeneration Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala 695012, India.,Inter University Centre for Biomedical Research and Super Speciality Hospital, Kottayam, Kerala 686009, India
| | - N Nimi
- Division of Tissue Engineering and Regeneration Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala 695012, India
| | - V P Sivadas
- Division of Tissue Engineering and Regeneration Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala 695012, India
| | - L P Merlin Rajesh Lal
- Division of Tissue Engineering and Regeneration Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala 695012, India
| | - Prabha D Nair
- Division of Tissue Engineering and Regeneration Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala 695012, India
| |
Collapse
|
171
|
Ohba S. Genome-scale actions of master regulators directing skeletal development. JAPANESE DENTAL SCIENCE REVIEW 2021; 57:217-223. [PMID: 34745394 PMCID: PMC8556520 DOI: 10.1016/j.jdsr.2021.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/14/2021] [Accepted: 10/10/2021] [Indexed: 11/03/2022] Open
Abstract
The mammalian skeleton develops through two distinct modes of ossification: intramembranous ossification and endochondral ossification. During the process of skeletal development, SRY-box containing gene 9 (Sox9), runt-related transcription factor 2 (Runx2), and Sp7 work as master transcription factors (TFs) or transcriptional regulators, underlying cell fate specification of the two distinct populations: bone-forming osteoblasts and cartilage-forming chondrocytes. In the past two decades, core transcriptional circuits underlying skeletal development have been identified mainly through mouse genetics and biochemical approaches. Recently emerging next-generation sequencer (NGS)-based studies have provided genome-scale views on the gene regulatory landscape programmed by the master TFs/transcriptional regulators. With particular focus on Sox9, Runx2, and Sp7, this review aims to discuss the gene regulatory landscape in skeletal development, which has been identified by genome-scale data, and provide future perspectives in this field.
Collapse
Affiliation(s)
- Shinsuke Ohba
- Department of Cell Biology, Institute of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| |
Collapse
|
172
|
Migale R, Neumann M, Lovell-Badge R. Long-Range Regulation of Key Sex Determination Genes. Sex Dev 2021; 15:360-380. [PMID: 34753143 DOI: 10.1159/000519891] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/26/2021] [Indexed: 11/19/2022] Open
Abstract
The development of sexually dimorphic gonads is a unique process that starts with the specification of the bipotential genital ridges and culminates with the development of fully differentiated ovaries and testes in females and males, respectively. Research on sex determination has been mostly focused on the identification of sex determination genes, the majority of which encode for proteins and specifically transcription factors such as SOX9 in the testes and FOXL2 in the ovaries. Our understanding of which factors may be critical for sex determination have benefited from the study of human disorders of sex development (DSD) and animal models, such as the mouse and the goat, as these often replicate the same phenotypes observed in humans when mutations or chromosomic rearrangements arise in protein-coding genes. Despite the advances made so far in explaining the role of key factors such as SRY, SOX9, and FOXL2 and the genes they control, what may regulate these factors upstream is not entirely understood, often resulting in the inability to correctly diagnose DSD patients. The role of non-coding DNA, which represents 98% of the human genome, in sex determination has only recently begun to be fully appreciated. In this review, we summarize the current knowledge on the long-range regulation of 2 important sex determination genes, SOX9 and FOXL2, and discuss the challenges that lie ahead and the many avenues of research yet to be explored in the sex determination field.
Collapse
|
173
|
Zfhx4 regulates endochondral ossification as the transcriptional platform of Osterix in mice. Commun Biol 2021; 4:1258. [PMID: 34732852 PMCID: PMC8566502 DOI: 10.1038/s42003-021-02793-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 10/18/2021] [Indexed: 11/08/2022] Open
Abstract
Endochondral ossification is regulated by transcription factors that include SRY-box transcription factor 9, runt-related protein 2 (Runx2), and Osterix. However, the sequential and harmonious regulation of the multiple steps of endochondral ossification is unclear. This study identified zinc finger homeodomain 4 (Zfhx4) as a crucial transcriptional partner of Osterix. We found that Zfhx4 was highly expressed in cartilage and that Zfhx4 deficient mice had reduced expression of matrix metallopeptidase 13 and inhibited calcification of cartilage matrices. These phenotypes were very similar to impaired chondrogenesis in Osterix deficient mice. Coimmunoprecipitation and immunofluorescence indicated a physical interaction between Zfhx4 and Osterix. Notably, Zfhx4 and Osterix double mutant mice showed more severe phenotype than Zfhx4 deficient mice. Additionally, Zfhx4 interacted with Runx2 that functions upstream of Osterix. Our findings suggest that Zfhx4 coordinates the transcriptional network of Osterix and, consequently, endochondral ossification.
Collapse
|
174
|
Ma L, Zhang R, Li D, Qiao T, Guo X. Fluoride regulates chondrocyte proliferation and autophagy via PI3K/AKT/mTOR signaling pathway. Chem Biol Interact 2021; 349:109659. [PMID: 34536393 DOI: 10.1016/j.cbi.2021.109659] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 08/17/2021] [Accepted: 09/14/2021] [Indexed: 01/08/2023]
Abstract
Fluorine is an essential trace element for human health. However, excessive fluoride intake causes skeletal fluorosis which affects cartilage development. Fluoride inhibited chondrocyte proliferation which is the initial and critical step of endochondral ossification, but the underlying mechanism has not been clearly illustrated. Mammalian target of rapamycin (mTOR) is an important protein kinase which modulates various cellular processes and is believed to be a central regulator of chondrocyte proliferation and autophagy. In this study, we explored the effect of fluoride on the proliferation and autophagy of chondrocytes and the regulatory role of mTOR signaling pathway. Our results suggested that NaF inhibited the protein expressions of proliferating cell nuclear antigen (PCNA) and pS6 in cultured fetal rat tibias. Furthermore, NaF significantly downregulated the expressions of mTOR signaling pathway-related genes, including PI3K, AKT, mTOR, 4EBP1 and S6K1 in mouse ATDC5 chondrogenic cell line. We also found that NaF increased autophagy in ATDC5 cells. The mRNA and protein levels of autophagy-related genes LC3, Beclin1 and p62 were significantly changed after NaF treatment. Further studies demonstrated that MHY1485, a small-molecular mTOR activator, totally reversed fluoride-induced promotion of autophagy. MHY1485 also recovered the downregulation of proliferative chondrocytes markers Sox9 and Type Ⅱ Collagen (Col2a1) induced by fluoride in ATDC5 cells. Taken together, our result demonstrate that fluoride suppressed proliferation and facilitated autophagy via PI3K/AKT/mTOR signaling pathway in chondrogenesis.
Collapse
Affiliation(s)
- Lan Ma
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Ruixue Zhang
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Demin Li
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Tingting Qiao
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Xiaoying Guo
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China.
| |
Collapse
|
175
|
Lange C, Madry H, Venkatesan JK, Schmitt G, Speicher-Mentges S, Zurakowski D, Menger MD, Laschke MW, Cucchiarini M. rAAV-Mediated sox9 Overexpression Improves the Repair of Osteochondral Defects in a Clinically Relevant Large Animal Model Over Time In Vivo and Reduces Perifocal Osteoarthritic Changes. Am J Sports Med 2021; 49:3696-3707. [PMID: 34643471 DOI: 10.1177/03635465211049414] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Gene transfer of the transcription factor SOX9 with clinically adapted recombinant adeno-associated virus (rAAV) vectors offers a powerful tool to durably enhance the repair process at sites of osteochondral injuries and counteract the development of perifocal osteoarthritis (OA) in the adjacent articular cartilage. PURPOSE To examine the ability of an rAAV sox9 construct to improve the repair of focal osteochondral defects and oppose perifocal OA development over time in a large translational model relative to control gene transfer. STUDY DESIGN Controlled laboratory study. METHODS Standardized osteochondral defects created in the knee joints of adult sheep were treated with rAAV-FLAG-hsox9 relative to control (reporter) rAAV-lacZ gene transfer. Osteochondral repair and degenerative changes in the adjacent cartilage were monitored using macroscopic, histological, immunohistological, and biochemical evaluations after 6 months. The microarchitecture of the subchondral bone was assessed by micro-computed tomography. RESULTS Effective, prolonged sox9 overexpression via rAAV was significantly achieved in the defects after 6 months versus rAAV-lacZ treatment. The application of rAAV-FLAG-hsox9 improved the individual parameters of defect filling, matrix staining, cellular morphology, defect architecture, surface architecture, subchondral bone, and tidemark as well as the overall score of cartilage repair in the defects compared with rAAV-lacZ. The overexpression of sox9 led to higher levels of proteoglycan production, stronger type II collagen deposition, and reduced type I collagen immunoreactivity in the sox9- versus lacZ-treated defects, together with decreased cell densities and DNA content. rAAV-FLAG-hsox9 enhanced semiquantitative histological subchondral bone repair, while the microstructure of the incompletely restored subchondral bone in the sox9 defects was not different from that in the lacZ defects. The articular cartilage adjacent to the sox9-treated defects showed reduced histological signs of perifocal OA changes versus rAAV-lacZ. CONCLUSION rAAV-mediated sox9 gene transfer enhanced osteochondral repair in sheep after 6 months and reduced perifocal OA changes. These results underline the potential of rAAV-FLAG-hsox9 as a therapeutic tool to treat cartilage defects and afford protection against OA. CLINICAL RELEVANCE The delivery of therapeutic rAAV sox9 to sites of focal injuries may offer a novel, convenient tool to enhance the repair of osteochondral defects involving both the articular cartilage and the underlying subchondral bone and provide a protective role by reducing the extent of perifocal OA.
Collapse
Affiliation(s)
- Cliff Lange
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg, Germany
| | - Jagadeesh K Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg, Germany
| | - Gertrud Schmitt
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg, Germany
| | | | - David Zurakowski
- Departments of Anesthesia and Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg, Germany
| |
Collapse
|
176
|
Moss JJ, Wirth M, Tooze SA, Lane JD, Hammond CL. Autophagy coordinates chondrocyte development and early joint formation in zebrafish. FASEB J 2021; 35:e22002. [PMID: 34708458 DOI: 10.1096/fj.202101167r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 12/16/2022]
Abstract
Autophagy is a catabolic process responsible for the removal of waste and damaged cellular components by lysosomal degradation. It plays a key role in fundamental cell processes, including ER stress mitigation, control of cell metabolism, and cell differentiation and proliferation, all of which are essential for cartilage cell (chondrocyte) development and survival, and for the formation of cartilage. Correspondingly, autophagy dysregulation has been implicated in several skeletal disorders such as osteoarthritis and osteoporosis. To test the requirement for autophagy during skeletal development in zebrafish, we generated an atg13 CRISPR knockout zebrafish line. This line showed a complete loss of atg13 expression, and restricted autophagic activity in vivo. In the absence of autophagy, chondrocyte maturation was accelerated, with chondrocytes exhibiting signs of premature hypertrophy. Focussing on the jaw element, autophagy disruption affected joint articulation causing restricted mouth opening. This gross behavioural phenotype corresponded with a failure to thrive, and death in homozygote atg13 nulls within 17 days. Taken together, our results are consistent with autophagy contributing to the timely regulation of chondrocyte maturation and for extracellular matrix formation.
Collapse
Affiliation(s)
- Joanna J Moss
- School of Biochemistry, University of Bristol, Bristol, UK.,School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Martina Wirth
- Molecular Cell Biology of Autophagy, The Francis Crick Institute, London, UK
| | - Sharon A Tooze
- Molecular Cell Biology of Autophagy, The Francis Crick Institute, London, UK
| | - Jon D Lane
- School of Biochemistry, University of Bristol, Bristol, UK
| | - Chrissy L Hammond
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| |
Collapse
|
177
|
Liu J, Zuo Q, Li Z, Chen J, Liu F. Trelagliptin ameliorates IL-1β-impaired chondrocyte function via the AMPK/SOX-9 pathway. Mol Immunol 2021; 140:70-76. [PMID: 34666245 DOI: 10.1016/j.molimm.2021.09.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 08/01/2021] [Accepted: 09/14/2021] [Indexed: 12/20/2022]
Abstract
Chondrocyte dysregulation plays a critical role in the development of osteoarthritis (OA). The pro-inflammatory cytokine interleukin-1β (IL-1β) activates chondrocytes and degrades the structural extracellular matrix (ECM). These events are the important mechanism of OA. Trelagliptin, a selective inhibitor of dipeptidyl Peptidase 4 (DPP-4) used for the treatment of type 2 diabetes mellitus (T2DM), has displayed a wide range of anti-inflammatory capacities. The effects of Trelagliptin in OA and chondrocytes have not been tested before. Here, we show that Trelagliptin mitigates IL-1β-induced production of inflammatory cytokines such as interleukin 6 (IL-6), interleukin 8 (IL-8), and tumor necrosis factor-alpha (TNF-α) in human chondrocytes. Trelagliptin ameliorates IL-1β-induced oxidative stress by reducing the generation of reactive oxygen species (ROS). Particularly, the presence of Trelagliptin prevents IL-1β-induced reduction of Acan genes and the protein Aggrecan. Moreover, we show that Trelagliptin restores IL-1β-induced reduction of SOX-9 and that the knockdown of SOX-9 abolishes the protective effects of Trelagliptin. Mechanistically, we demonstrate that AMPK is required for the amelioration of Trelagliptin on SOX-9- reduction by IL-1β. Collectively, our study demonstrates that the DPP-4 inhibitor Trelagliptin has a protective effect on chondrocyte function. Trelagliptin may have the potential role to antagonize chondrocyte-derived inflammation in OA.
Collapse
Affiliation(s)
- Jiuxiang Liu
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Nanjing Medical University (Jiangsu Province Hospital), Nanjing, Jiangsu, 210029, China
| | - Qiang Zuo
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Nanjing Medical University (Jiangsu Province Hospital), Nanjing, Jiangsu, 210029, China
| | - Zhi Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Nanjing Medical University (Jiangsu Province Hospital), Nanjing, Jiangsu, 210029, China
| | - Jiangqi Chen
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Nanjing Medical University (Jiangsu Province Hospital), Nanjing, Jiangsu, 210029, China
| | - Feng Liu
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Nanjing Medical University (Jiangsu Province Hospital), Nanjing, Jiangsu, 210029, China.
| |
Collapse
|
178
|
Pan D, Qian B, Zhao D, Yao B. Nfib promotes chondrocyte proliferation and inhibits differentiation by mildly regulating Sox9 and its downstream genes. Mol Biol Rep 2021; 48:7487-7497. [PMID: 34651294 DOI: 10.1007/s11033-021-06767-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 09/15/2021] [Indexed: 01/18/2023]
Abstract
BACKGROUND Chondrocyte proliferation and differentiation play pivotal roles in regulating cartilage formation, endochondral bone formation, and repair. Cartilage damage and underdevelopment may cause severe joint diseases. Various transcription factors regulate cartilage development. Nuclear factor 1 B (Nfib) is a transcription factor that plays a regulatory role in various organs. However, the effect and mechanism of Nfib on the proliferation and differentiation of chondrocytes in cartilage are still largely unknown. METHODS AND RESULTS In the present study, we investigated the gene expression patterns in primary chondrocytes with Nfib overexpression or silencing by RNA sequencing (RNA-seq) technology. The results showed that Nfib overexpression significantly up-regulated genes that are related to chondrocyte proliferation and extracellular matrix (ECM) synthesis and significantly down-regulated genes related to chondrocyte differentiation and ECM degradation. However, with Nfib silencing, the genes involved in promoting chondrocyte differentiation were significantly up-regulated, whereas those involved in promoting chondrocyte proliferation were significantly down-regulated. Furthermore, quantitative real-time PCR (qRT-PCR), western blot, alcian blue staining and immunofluorescence staining assays further confirmed that Nfib potentially promotes chondrocyte proliferation and extracellular synthesis but inhibits differentiation. CONCLUSIONS The molecular mechanism of Nfib in promoting chondrocyte proliferation and inhibiting differentiation was probably achieved by stimulating Sox9 and its downstream genes. Thus, this study adds new insights regarding the underlying molecular mechanism of transcriptional regulation in cartilage.
Collapse
Affiliation(s)
- Daian Pan
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Benxin Qian
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Daqing Zhao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China.
| | - Baojin Yao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China.
| |
Collapse
|
179
|
McNamara JT, Huntington KE, Borys S, Jayasuriya CT, Brossay L. SHP-2 deletion in CD4Cre expressing chondrocyte precursors leads to tumor development with wrist tropism. Sci Rep 2021; 11:20006. [PMID: 34625577 PMCID: PMC8501018 DOI: 10.1038/s41598-021-99339-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 09/20/2021] [Indexed: 11/16/2022] Open
Abstract
Due to redundancy with other tyrosine phosphatases, the ubiquitously expressed tyrosine phosphatase SHP-2 (encoded by Ptpn11) is not required for T cell development. However, Ptpn11 gene deletion driven by CD4 Cre recombinase leads to cartilage tumors in the wrist. Using a fate mapping system, we demonstrate that wrist tumor development correlates with increased frequency and numbers of non-hematopoietic lineage negative CD45 negative cells with a bone chondrocyte stromal cell precursor cell (BCSP) phenotype. Importantly, the BCSP subset has a history of CD4 expression and a marked wrist location tropism, explaining why the wrist is the main site of tumor development. Mechanistically, we found that in SHP-2 absence, SOX-9 is no longer regulated, leading to an uncontrolled proliferation of the BCSP subset. Altogether, these results identify a unique subset of chondrocyte precursors tightly regulated by SHP-2. These findings underscore the need for the development of methods to therapeutically target this subset of cells, which could potentially have an impact on treatment of SHP-2 dysfunction linked debilitating diseases.
Collapse
Affiliation(s)
- Jeffrey T McNamara
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University Alpert Medical School, Providence, RI, 02912, USA
| | - Kelsey E Huntington
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University Alpert Medical School, Providence, RI, 02912, USA
| | - Samantha Borys
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University Alpert Medical School, Providence, RI, 02912, USA
| | - Chathuraka T Jayasuriya
- Department of Orthopaedics, Rhode Island Hospital and Brown University Alpert Medical School, Providence, RI, USA
| | - Laurent Brossay
- Division of Biology and Medicine, Department of Molecular Microbiology and Immunology, Brown University Alpert Medical School, Providence, RI, 02912, USA.
| |
Collapse
|
180
|
Oncogenic role of SOX9-DHCR24-cholesterol biosynthesis axis in IGH-BCL2 positive diffuse large B-cell lymphomas. Blood 2021; 139:73-86. [PMID: 34624089 PMCID: PMC8740888 DOI: 10.1182/blood.2021012327] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/29/2021] [Indexed: 11/21/2022] Open
Abstract
SOX9 plays an oncogenic role in germinal center B-cell type, IGH-BCL2+ DLBCL, by promoting cell proliferation and inhibiting apoptosis. SOX9 drives lymphomagenesis through upregulation of DHCR24, the key final enzyme in the cholesterol biosynthesis pathway.
Although oncogenicity of the stem cell regulator SOX9 has been implicated in many solid tumors, its role in lymphomagenesis remains largely unknown. In this study, SOX9 was overexpressed preferentially in a subset of diffuse large B-cell lymphomas (DLBCLs) that harbor IGH-BCL2 translocations. SOX9 positivity in DLBCL correlated with an advanced stage of disease. Silencing of SOX9 decreased cell proliferation, induced G1/S arrest, and increased apoptosis of DLBCL cells, both in vitro and in vivo. Whole-transcriptome analysis and chromatin immunoprecipitation–sequencing assays identified DHCR24, a terminal enzyme in cholesterol biosynthesis, as a direct target of SOX9, which promotes cholesterol synthesis by increasing DHCR24 expression. Enforced expression of DHCR24 was capable of rescuing the phenotypes associated with SOX9 knockdown in DLBCL cells. In models of DLBCL cell line xenografts, SOX9 knockdown resulted in a lower DHCR24 level, reduced cholesterol content, and decreased tumor load. Pharmacological inhibition of cholesterol synthesis also inhibited DLBCL xenograft tumorigenesis, the reduction of which is more pronounced in DLBCL cell lines with higher SOX9 expression, suggesting that it may be addicted to cholesterol. In summary, our study demonstrated that SOX9 can drive lymphomagenesis through DHCR24 and the cholesterol biosynthesis pathway. This SOX9-DHCR24-cholesterol biosynthesis axis may serve as a novel treatment target for DLBCLs.
Collapse
|
181
|
Asmussen NC, Cohen DJ, Lin Z, McClure MJ, Boyan BD, Schwartz Z. Specific MicroRNAs Found in Extracellular Matrix Vesicles Regulate Proliferation and Differentiation in Growth Plate Chondrocytes. Calcif Tissue Int 2021; 109:455-468. [PMID: 33950267 DOI: 10.1007/s00223-021-00855-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/19/2021] [Indexed: 10/21/2022]
Abstract
Matrix vesicles (MVs) are extracellular organelles produced by growth plate cartilage cells in a zone-specific manner. MVs are similar in size to exosomes, but they are tethered to the extracellular matrix (ECM) via integrins. Originally associated with matrix calcification, studies now show that they contain matrix processing enzymes and microRNA that are specific to their zone of maturation. MVs produced by costochondral cartilage resting zone (RC) chondrocytes are enriched in microRNA 503 whereas those produced by growth zone (GC) chondrocytes are enriched in microRNA 122. MVs are packaged by chondrocytes under hormonal and factor regulation and release of their contents into the ECM is also under hormonal control, suggesting that their microRNA might have a regulatory role in growth plate proliferation and maturation. To test this, we selected a subset of these enriched microRNAs and transfected synthetic mimics back into RC and GC cells. Transfecting growth plate chondrocytes with select microRNA produced a broad range of phenotypic responses indicating that MV-based microRNAs are involved in the regulation of these cells. Specifically, microRNA 122 drives both RC and GC cells toward a proliferative phenotype, stabilizes the matrix and inhibits differentiation whereas microRNA 22 exerts control over regulatory factor production. This study demonstrates the strong regulatory capability possessed by unique MV enriched microRNAs on growth plate chondrocytes and their potential for use as therapeutic agents.
Collapse
Affiliation(s)
- Niels C Asmussen
- School of Integrative Life Sciences, Virginia Commonwealth University, Richmond, VA, 23284, USA
| | - David J Cohen
- College of Engineering, Virginia Commonwealth University, 601 W. Main Street, Richmond, VA, 23284, USA
| | - Zhao Lin
- School of Dentistry, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Michael J McClure
- College of Engineering, Virginia Commonwealth University, 601 W. Main Street, Richmond, VA, 23284, USA
| | - Barbara D Boyan
- College of Engineering, Virginia Commonwealth University, 601 W. Main Street, Richmond, VA, 23284, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| | - Zvi Schwartz
- College of Engineering, Virginia Commonwealth University, 601 W. Main Street, Richmond, VA, 23284, USA
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| |
Collapse
|
182
|
Abraham SP, Nita A, Krejci P, Bosakova M. Cilia kinases in skeletal development and homeostasis. Dev Dyn 2021; 251:577-608. [PMID: 34582081 DOI: 10.1002/dvdy.426] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 11/08/2022] Open
Abstract
Primary cilia are dynamic compartments that regulate multiple aspects of cellular signaling. The production, maintenance, and function of cilia involve more than 1000 genes in mammals, and their mutations disrupt the ciliary signaling which manifests in a plethora of pathological conditions-the ciliopathies. Skeletal ciliopathies are genetic disorders affecting the development and homeostasis of the skeleton, and encompass a broad spectrum of pathologies ranging from isolated polydactyly to lethal syndromic dysplasias. The recent advances in forward genetics allowed for the identification of novel regulators of skeletogenesis, and revealed a growing list of ciliary proteins that are critical for signaling pathways implicated in bone physiology. Among these, a group of protein kinases involved in cilia assembly, maintenance, signaling, and disassembly has emerged. In this review, we summarize the functions of cilia kinases in skeletal development and disease, and discuss the available and upcoming treatment options.
Collapse
Affiliation(s)
- Sara P Abraham
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Alexandru Nita
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Pavel Krejci
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Institute of Animal Physiology and Genetics of the CAS, Brno, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Michaela Bosakova
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Institute of Animal Physiology and Genetics of the CAS, Brno, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| |
Collapse
|
183
|
Shimada M, Kanno N, Ichinohe T, Suzuki S, Harada Y, Hara Y. Prophylactic Efficacy of Tibial Plateau Levelling Osteotomy for a Canine Model with Experimentally Induced Degeneration of the Cranial Cruciate Ligament. Vet Comp Orthop Traumatol 2021; 35:18-25. [PMID: 34547788 DOI: 10.1055/s-0041-1735318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE The aim of this study was to clarify the histological effects of tibial plateau levelling osteotomy on cranial cruciate ligament degeneration induced by excessive tibial plateau angle. STUDY DESIGN Five female Beagles were used to bilaterally create excessive tibial plateau angle models surgically. A second tibial plateau levelling osteotomy was performed 11 months after the first surgery on the right stifle (tibial plateau levelling osteotomy group), and a sham operation that did not change the tibial plateau angle was performed on the left stifle (excessive tibial plateau angle group). At 6 months after the second surgery, the dogs were euthanatized. The cranial cruciate ligament was stained with haematoxylin-eosin to assess the cell density, Alcian-Blue to assess proteoglycans and Elastica-Eosin to assess elastic fibres, and immunohistochemically stained to assess type I (COL1) and type II collagen and SRY-type HMG box 9 (SOX9) expression. RESULTS In each group, the cranial cruciate ligament degeneration, especially on the tibial side, including the presence of Alcian-Blue- and Elastica-Eosin-positive regions, decreased in COL1-positive regions, and enhancement of SOX9 expression was observed. Besides, compared with the tibial plateau levelling osteotomy group, the excessive tibial plateau angle group showed increases in Alcian-Blue- and Elastica-Eosin-positive regions and a decrease in the COL1-positive regions. CONCLUSION The results suggested that excessive tibial plateau angle-induced cranial cruciate ligament degeneration can be suppressed by reducing the biomechanical load on the cranial cruciate ligament by performing tibial plateau levelling osteotomy.
Collapse
Affiliation(s)
- Masakazu Shimada
- Division of Veterinary Surgery, Department of Veterinary Science, Faculty of Veterinary Medicine, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| | - Nobuo Kanno
- Division of Veterinary Surgery, Department of Veterinary Science, Faculty of Veterinary Medicine, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| | - Tom Ichinohe
- Division of Veterinary Surgery, Department of Veterinary Science, Faculty of Veterinary Medicine, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan.,Veterinary Teaching Hospital, Azabu University, Chuou-ku, Sagamihara-shi, Kanagawa, Japan
| | - Shuji Suzuki
- Division of Veterinary Surgery, Department of Veterinary Science, Faculty of Veterinary Medicine, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| | - Yasuji Harada
- Division of Veterinary Surgery, Department of Veterinary Science, Faculty of Veterinary Medicine, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| | - Yasushi Hara
- Division of Veterinary Surgery, Department of Veterinary Science, Faculty of Veterinary Medicine, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| |
Collapse
|
184
|
Kuhlmann C, Schenck TL, Aszodi A, Giunta RE, Wiggenhauser PS. Zone-Dependent Architecture and Biochemical Composition of Decellularized Porcine Nasal Cartilage Modulate the Activity of Adipose Tissue-Derived Stem Cells in Cartilage Regeneration. Int J Mol Sci 2021; 22:ijms22189917. [PMID: 34576079 PMCID: PMC8470846 DOI: 10.3390/ijms22189917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/27/2021] [Accepted: 09/03/2021] [Indexed: 01/22/2023] Open
Abstract
Previous anatomical studies have shown different functional zones in human nasal septal cartilage (NC). These zones differ in respect to histological architecture and biochemical composition. The aim of this study was to investigate the influence of these zones on the fate of stem cells from a regenerative perspective. Therefore, decellularized porcine septal cartilage was prepared and subjected to histological assessment to demonstrate its equivalence to human cartilage. Decellularized porcine NC (DPNC) exposed distinct surfaces depending on two different histological zones: the outer surface (OS), which is equivalent to the superficial zone, and the inner surface (IS), which is equivalent to the central zone. Human adipose tissue-derived stem cells (ASCs) were isolated from the abdominal fat tissue of five female patients and were seeded on the IS and OS of DPNC, respectively. Cell seeding efficiency (CSE), vitality, proliferation, migration, the production of sulfated glycosaminoglycans (sGAG) and chondrogenic differentiation capacity were evaluated by histological staining (DAPI, Phalloidin, Live-Dead), biochemical assays (alamarBlue®, PicoGreen®, DMMB) and the quantification of gene expression (qPCR). Results show that cell vitality and CSE were not influenced by DPNC zones. ASCs, however, showed a significantly higher proliferation and elevated expression of early chondrogenic differentiation, as well as fibrocartilage markers, on the OS. On the contrary, there was a significantly higher upregulation of hypertrophy marker MMP13 (p < 0.0001) and GAG production (p = 0.0105) on the IS, whereas cell invasion into the three-dimensional DPNC was higher in comparison to the OS. We conclude that the zonal-dependent distinct architecture and composition of NC modulates activities of ASCs seeded on DPNC. These findings might be used for engineering of cartilage substitutes needed in facial reconstructive surgery that yield an equivalent histological and functional structure, such as native NC.
Collapse
Affiliation(s)
- Constanze Kuhlmann
- Department of Hand, Plastic and Aesthetic Surgery, LMU Klinikum, University Hospital, LMU Munich, Ziemsenstrasse 5, 80336 Munich, Germany; (C.K.); (T.L.S.); (R.E.G.)
- Laboratory of Cartilage Development, Diseases and Regeneration, Department for Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), University Hospital, LMU Munich, Fraunhoferstrasse 20, 82152 Planegg, Germany;
| | - Thilo L. Schenck
- Department of Hand, Plastic and Aesthetic Surgery, LMU Klinikum, University Hospital, LMU Munich, Ziemsenstrasse 5, 80336 Munich, Germany; (C.K.); (T.L.S.); (R.E.G.)
| | - Attila Aszodi
- Laboratory of Cartilage Development, Diseases and Regeneration, Department for Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), University Hospital, LMU Munich, Fraunhoferstrasse 20, 82152 Planegg, Germany;
| | - Riccardo E. Giunta
- Department of Hand, Plastic and Aesthetic Surgery, LMU Klinikum, University Hospital, LMU Munich, Ziemsenstrasse 5, 80336 Munich, Germany; (C.K.); (T.L.S.); (R.E.G.)
| | - Paul Severin Wiggenhauser
- Department of Hand, Plastic and Aesthetic Surgery, LMU Klinikum, University Hospital, LMU Munich, Ziemsenstrasse 5, 80336 Munich, Germany; (C.K.); (T.L.S.); (R.E.G.)
- Correspondence:
| |
Collapse
|
185
|
Mok CH, MacLeod JN. Kinetics of Gene Expression Changes in Equine Fetal Interzone and Anlagen Cells Over 14 Days of Induced Chondrogenesis. Front Vet Sci 2021; 8:722324. [PMID: 34434986 PMCID: PMC8380811 DOI: 10.3389/fvets.2021.722324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/13/2021] [Indexed: 11/13/2022] Open
Abstract
Within developing synovial joints, interzone and anlagen cells progress through divergent chondrogenic pathways to generate stable articular cartilage and transient hypertrophic anlagen cartilage, respectively. Understanding the comparative cell biology between interzone and anlagen cells may provide novel insights into emergent cell-based therapies to support articular cartilage regeneration. The aim of this study was to assess the kinetics of gene expression profiles in these skeletal cell lines after inducing chondrogenesis in culture. Interzone and anlagen cells from seven equine fetuses were isolated and grown in a TGF-β1 chondrogenic inductive medium. Total RNA was isolated at ten time points (0, 1.5, 3, 6, 12, 24, 48, 96, 168, and 336 h), and gene expression for 93 targeted gene loci was measured in a microfluidic RT-qPCR system. Differential transcriptional responses were observed as early as 1.5 h after the initiation of chondrogenesis. Genes with functional annotations that include transcription regulation responded to the chondrogenic stimulation earlier (1.5–96 h) than genes involved in signal transduction (1.5–336 h) and the extracellular matrix biology (3–336 h). Between interzone and anlagen cell cultures, expression levels of 73 out of the 93 targeted genes were not initially different at 0 h, but 47 out of the 73 genes became differentially expressed under the chondrogenic stimulation. While interzone and anlagen cells are both chondrogenic, they display clear differences in response to the same TGF-β1 chondrogenic stimulation. This study provides new molecular insight into a timed sequence of the divergent developmental fates of interzone and anlagen cells in culture over 14 days.
Collapse
Affiliation(s)
- Chan Hee Mok
- Department of Veterinary Science, Gluck Equine Research Center, University of Kentucky, Lexington, KY, United States
| | - James N MacLeod
- Department of Veterinary Science, Gluck Equine Research Center, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
186
|
Mizoguchi T, Ono N. The diverse origin of bone-forming osteoblasts. J Bone Miner Res 2021; 36:1432-1447. [PMID: 34213032 PMCID: PMC8338797 DOI: 10.1002/jbmr.4410] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/29/2021] [Accepted: 06/29/2021] [Indexed: 12/17/2022]
Abstract
Osteoblasts are the only cells that can give rise to bones in vertebrates. Thus, one of the most important functions of these metabolically active cells is mineralized matrix production. Because osteoblasts have a limited lifespan, they must be constantly replenished by preosteoblasts, their immediate precursors. Because disruption of the regulation of bone-forming osteoblasts results in a variety of bone diseases, a better understanding of the origin of these cells by defining the mechanisms of bone development, remodeling, and regeneration is central to the development of novel therapeutic approaches. In recent years, substantial new insights into the origin of osteoblasts-largely owing to rapid technological advances in murine lineage-tracing approaches and other single-cell technologies-have been obtained. Collectively, these findings indicate that osteoblasts involved in bone formation under various physiological, pathological, and therapeutic conditions can be obtained from numerous sources. The origins of osteoblasts include, but are not limited to, chondrocytes in the growth plate, stromal cells in the bone marrow, quiescent bone-lining cells on the bone surface, and specialized fibroblasts in the craniofacial structures, such as sutures and periodontal ligaments. Because osteoblasts can be generated from local cellular sources, bones can flexibly respond to regenerative and anabolic cues. However, whether osteoblasts derived from different cellular sources have distinct functions remains to be investigated. Currently, we are at the initial stage to aptly unravel the incredible diversity of the origins of bone-forming osteoblasts. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
| | - Noriaki Ono
- University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, USA
| |
Collapse
|
187
|
Almubarak A, Lavy R, Srnic N, Hu Y, Maripuri DP, Kume T, Berry FB. Loss of Foxc1 and Foxc2 function in chondroprogenitor cells disrupts endochondral ossification. J Biol Chem 2021; 297:101020. [PMID: 34331943 PMCID: PMC8383119 DOI: 10.1016/j.jbc.2021.101020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 07/12/2021] [Accepted: 07/27/2021] [Indexed: 11/23/2022] Open
Abstract
Endochondral ossification initiates the growth of the majority of the mammalian skeleton and is tightly controlled through gene regulatory networks. The forkhead box transcription factors Foxc1 and Foxc2 regulate aspects of osteoblast function in the formation of the skeleton, but their roles in chondrocytes to control endochondral ossification are less clear. Here, we demonstrate that Foxc1 expression is directly regulated by the activity of SRY (sex-determining region Y)-box 9, one of the earliest transcription factors to specify the chondrocyte lineage. Moreover, we demonstrate that elevated expression of Foxc1 promotes chondrocyte differentiation in mouse embryonic stem cells and loss of Foxc1 function inhibits chondrogenesis in vitro. Using chondrocyte-targeted deletion of Foxc1 and Foxc2 in mice, we reveal a role for these factors in chondrocyte differentiation in vivo. Loss of both Foxc1 and Foxc2 caused a general skeletal dysplasia predominantly affecting the vertebral column. The long bones of the limbs were smaller, mineralization was reduced, and organization of the growth plate was disrupted; in particular, the stacked columnar organization of the proliferative chondrocyte layer was reduced in size and cell proliferation was decreased. Differential gene expression analysis indicated disrupted expression patterns of chondrogenesis and ossification genes throughout the entire process of endochondral ossification in chondrocyte-specific Foxc1/Foxc2 KO embryos. Our results suggest that Foxc1 and Foxc2 are required for normal chondrocyte differentiation and function, as loss of both genes results in disorganization of the growth plate, reduced chondrocyte proliferation, and delays in chondrocyte hypertrophy that prevents ossification of the skeleton.
Collapse
Affiliation(s)
- Asra Almubarak
- Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| | - Rotem Lavy
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Nikola Srnic
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Yawen Hu
- Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| | | | - Tsutomo Kume
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Department of Medicine, Northwestern University, Chicago, IL, USA
| | - Fred B Berry
- Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada; Department of Surgery, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
188
|
Lan X, Liang Y, Erkut EJN, Kunze M, Mulet-Sierra A, Gong T, Osswald M, Ansari K, Seikaly H, Boluk Y, Adesida AB. Bioprinting of human nasoseptal chondrocytes-laden collagen hydrogel for cartilage tissue engineering. FASEB J 2021; 35:e21191. [PMID: 33595884 DOI: 10.1096/fj.202002081r] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/18/2020] [Accepted: 10/29/2020] [Indexed: 12/23/2022]
Abstract
Skin cancer patients often have tumorigenic lesions on their noses. Surgical resection of the lesions often results in nasal cartilage removal. Cartilage grafts taken from other anatomical sites are used for the surgical reconstruction of the nasal cartilage, but donor-site morbidity is a common problem. Autologous tissue-engineered nasal cartilage grafts can mitigate the problem, but commercially available scaffolds define the shape and sizes of the engineered grafts during tissue fabrication. Moreover, the engineered grafts suffer from the inhomogeneous distribution of the functional matrix of cartilage. Advances in 3D bioprinting technology offer the opportunity to engineer cartilages with customizable dimensions and anatomically shaped configurations without the inhomogeneous distribution of cartilage matrix. Here, we report the fidelity of Freeform Reversible Embedding of Suspended Hydrogel (FRESH) bioprinting as a strategy to generate customizable and homogenously distributed functional cartilage matrix engineered nasal cartilage. Using FRESH and in vitro chondrogenesis, we have fabricated tissue-engineered nasal cartilage from combining bovine type I collagen hydrogel and human nasoseptal chondrocytes. The engineered nasal cartilage constructs displayed molecular, biochemical and histological characteristics akin to native human nasal cartilage.
Collapse
Affiliation(s)
- Xiaoyi Lan
- Department of Civil and Environmental Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada
| | - Yan Liang
- Department of Surgery, Divisions of Orthopedic Surgery & Surgical Research, University of Alberta, Edmonton, AB, Canada
| | - Esra J N Erkut
- Department of Surgery, Divisions of Orthopedic Surgery & Surgical Research, University of Alberta, Edmonton, AB, Canada
| | - Melanie Kunze
- Department of Surgery, Divisions of Orthopedic Surgery & Surgical Research, University of Alberta, Edmonton, AB, Canada
| | - Aillette Mulet-Sierra
- Department of Surgery, Divisions of Orthopedic Surgery & Surgical Research, University of Alberta, Edmonton, AB, Canada
| | - Tianxing Gong
- Department of Surgery, Divisions of Orthopedic Surgery & Surgical Research, University of Alberta, Edmonton, AB, Canada
| | - Martin Osswald
- Institute for Reconstructive Sciences in Medicine (iRSM), Misericordia Community Hospital, Edmonton, AB, Canada.,Department of Surgery, Division of Otolaryngology, University of Alberta, Edmonton, AB, Canada
| | - Khalid Ansari
- Department of Surgery, Division of Otolaryngology, University of Alberta, Edmonton, AB, Canada
| | - Hadi Seikaly
- Department of Surgery, Division of Otolaryngology, University of Alberta, Edmonton, AB, Canada
| | - Yaman Boluk
- Department of Civil and Environmental Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada
| | - Adetola B Adesida
- Department of Surgery, Divisions of Orthopedic Surgery & Surgical Research, University of Alberta, Edmonton, AB, Canada.,Department of Surgery, Division of Otolaryngology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
189
|
Park S, Bello A, Arai Y, Ahn J, Kim D, Cha KY, Baek I, Park H, Lee SH. Functional Duality of Chondrocyte Hypertrophy and Biomedical Application Trends in Osteoarthritis. Pharmaceutics 2021; 13:pharmaceutics13081139. [PMID: 34452101 PMCID: PMC8400409 DOI: 10.3390/pharmaceutics13081139] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 11/16/2022] Open
Abstract
Chondrocyte hypertrophy is one of the key indicators in the progression of osteoarthritis (OA). However, compared with other OA indications, such as cartilage collapse, sclerosis, inflammation, and protease activation, the mechanisms by which chondrocyte hypertrophy contributes to OA remain elusive. As the pathological processes in the OA cartilage microenvironment, such as the alterations in the extracellular matrix, are initiated and dictated by the physiological state of the chondrocytes, in-depth knowledge of chondrocyte hypertrophy is necessary to enhance our understanding of the disease pathology and develop therapeutic agents. Chondrocyte hypertrophy is a factor that induces OA progression; it is also a crucial factor in the endochondral ossification. This review elaborates on this dual functionality of chondrocyte hypertrophy in OA progression and endochondral ossification through a description of the characteristics of various genes and signaling, their mechanism, and their distinguishable physiological effects. Chondrocyte hypertrophy in OA progression leads to a decrease in chondrogenic genes and destruction of cartilage tissue. However, in endochondral ossification, it represents an intermediate stage at the process of differentiation of chondrocytes into osteogenic cells. In addition, this review describes the current therapeutic strategies and their mechanisms, involving genes, proteins, cytokines, small molecules, three-dimensional environments, or exosomes, against the OA induced by chondrocyte hypertrophy. Finally, this review proposes that the contrasting roles of chondrocyte hypertrophy are essential for both OA progression and endochondral ossification, and that this cellular process may be targeted to develop OA therapeutics.
Collapse
Affiliation(s)
- Sunghyun Park
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea
| | - Alvin Bello
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
- School of Integrative Engineering, Chung-ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Korea;
| | - Yoshie Arai
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
| | - Jinsung Ahn
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
| | - Dohyun Kim
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
| | - Kyung-Yup Cha
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
| | - Inho Baek
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
| | - Hansoo Park
- School of Integrative Engineering, Chung-ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Korea;
| | - Soo-Hong Lee
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
- Correspondence: ; Tel.: +82-31-961-5153; Fax: +82-31-961-5108
| |
Collapse
|
190
|
Sun X, Zhang J, Li Y, Ren W, Wang L. Etomidate ameliorated advanced glycation end-products (AGEs)-induced reduction of extracellular matrix genes expression in chondrocytes. Bioengineered 2021; 12:4191-4200. [PMID: 34308765 PMCID: PMC8806553 DOI: 10.1080/21655979.2021.1951926] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Osteoarthritis (OA) is a rheumatic disease common in the elderly. AGEs are the end products of glycation reactions and play an important role in the development of OA. Etomidate is a general anesthesia-inducing agent recently reported to exert significant anti-inflammatory effects. The present study aims to explore the protective effect of Etomidate against advanced glycation end-products (AGEs)-induced reduction of extracellular matrix gene expression in chondrocytes. In the present study, we found that AGEs significantly reduced the expression of Collagen II (COL2A1) and Aggrecan (ACAN) at the gene level. Furthermore, AGEs inhibited the expression of SRY-related high mobility group-box gene 9 (SOX-9), promoting the expression of COL2A1 and ACAN. COL2A1, ACAN, and SOX-9 in chondrocytes were significantly elevated by treatment with Etomidate alone. Consistently, Etomidate ameliorated AGEs-induced downregulation of COL2A1, ACAN, and SOX-9 in a dose-dependent manner. Importantly, we found that knockdown of SOX-9 eliminated the beneficial effects of Etomidate against AGEs-induced decrease in COL2A1 and ACAN genes. Based on these findings, we demonstrated that Etomidate could ameliorate AGEs-induced reduction of extracellular matrix gene expression in chondrocytes by upregulating SOX-9.
Collapse
Affiliation(s)
- Xiaohua Sun
- Department of Anesthesiology, Outpatient and Emergency, Tianjin Hospital, Tianjin, China
| | - Jizheng Zhang
- Department of Anesthesiology, Outpatient and Emergency, Tianjin Hospital, Tianjin, China
| | - Yi Li
- Department of Anesthesiology, Outpatient and Emergency, Tianjin Hospital, Tianjin, China
| | - Wanlu Ren
- Department of Anesthesiology, Outpatient and Emergency, Tianjin Hospital, Tianjin, China
| | - Lijun Wang
- Department of Anus& Intestine Surgery, Tianjin Hospital, Tianjin, China
| |
Collapse
|
191
|
Fabik J, Psutkova V, Machon O. The Mandibular and Hyoid Arches-From Molecular Patterning to Shaping Bone and Cartilage. Int J Mol Sci 2021; 22:7529. [PMID: 34299147 PMCID: PMC8303155 DOI: 10.3390/ijms22147529] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 12/16/2022] Open
Abstract
The mandibular and hyoid arches collectively make up the facial skeleton, also known as the viscerocranium. Although all three germ layers come together to assemble the pharyngeal arches, the majority of tissue within viscerocranial skeletal components differentiates from the neural crest. Since nearly one third of all birth defects in humans affect the craniofacial region, it is important to understand how signalling pathways and transcription factors govern the embryogenesis and skeletogenesis of the viscerocranium. This review focuses on mouse and zebrafish models of craniofacial development. We highlight gene regulatory networks directing the patterning and osteochondrogenesis of the mandibular and hyoid arches that are actually conserved among all gnathostomes. The first part of this review describes the anatomy and development of mandibular and hyoid arches in both species. The second part analyses cell signalling and transcription factors that ensure the specificity of individual structures along the anatomical axes. The third part discusses the genes and molecules that control the formation of bone and cartilage within mandibular and hyoid arches and how dysregulation of molecular signalling influences the development of skeletal components of the viscerocranium. In conclusion, we notice that mandibular malformations in humans and mice often co-occur with hyoid malformations and pinpoint the similar molecular machinery controlling the development of mandibular and hyoid arches.
Collapse
Affiliation(s)
- Jaroslav Fabik
- Department of Developmental Biology, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic; (J.F.); (V.P.)
- Department of Cell Biology, Faculty of Science, Charles University, 12800 Prague, Czech Republic
| | - Viktorie Psutkova
- Department of Developmental Biology, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic; (J.F.); (V.P.)
- Department of Cell Biology, Faculty of Science, Charles University, 12800 Prague, Czech Republic
| | - Ondrej Machon
- Department of Developmental Biology, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic; (J.F.); (V.P.)
| |
Collapse
|
192
|
Rauch A, Mandrup S. Transcriptional networks controlling stromal cell differentiation. Nat Rev Mol Cell Biol 2021; 22:465-482. [PMID: 33837369 DOI: 10.1038/s41580-021-00357-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2021] [Indexed: 02/02/2023]
Abstract
Stromal progenitors are found in many different tissues, where they play an important role in the maintenance of tissue homeostasis owing to their ability to differentiate into parenchymal cells. These progenitor cells are differentially pre-programmed by their tissue microenvironment but, when cultured and stimulated in vitro, these cells - commonly referred to as mesenchymal stromal cells (MSCs) - exhibit a marked plasticity to differentiate into many different cell lineages. Loss-of-function studies in vitro and in vivo have uncovered the involvement of specific signalling pathways and key transcriptional regulators that work in a sequential and coordinated fashion to activate lineage-selective gene programmes. Recent advances in omics and single-cell technologies have made it possible to obtain system-wide insights into the gene regulatory networks that drive lineage determination and cell differentiation. These insights have important implications for the understanding of cell differentiation, the contribution of stromal cells to human disease and for the development of cell-based therapeutic applications.
Collapse
Affiliation(s)
- Alexander Rauch
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, Odense, Denmark. .,Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark.
| | - Susanne Mandrup
- Center for Functional Genomics and Tissue Plasticity, Functional Genomics & Metabolism Research Unit, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
193
|
Kishimoto K, Morimoto M. Mammalian tracheal development and reconstruction: insights from in vivo and in vitro studies. Development 2021; 148:dev198192. [PMID: 34228796 PMCID: PMC8276987 DOI: 10.1242/dev.198192] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The trachea delivers inhaled air into the lungs for gas exchange. Anomalies in tracheal development can result in life-threatening malformations, such as tracheoesophageal fistula and tracheomalacia. Given the limitations of current therapeutic approaches, development of technologies for the reconstitution of a three-dimensional trachea from stem cells is urgently required. Recently, single-cell sequencing technologies and quantitative analyses from cell to tissue scale have been employed to decipher the cellular basis of tracheal morphogenesis. In this Review, recent advances in mammalian tracheal development and the generation of tracheal tissues from pluripotent stem cells are summarized.
Collapse
Affiliation(s)
- Keishi Kishimoto
- Laboratory for Lung Development and Regeneration, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe 650-0047, Japan
- RIKEN BDR–CuSTOM Joint Laboratory, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Center for Stem Cell & Organoid Medicine (CuSTOM), Perinatal Institute, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Mitsuru Morimoto
- Laboratory for Lung Development and Regeneration, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe 650-0047, Japan
- RIKEN BDR–CuSTOM Joint Laboratory, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
194
|
Yadav PS, Feng S, Cong Q, Kim H, Liu Y, Yang Y. Stat3 loss in mesenchymal progenitors causes Job syndrome-like skeletal defects by reducing Wnt/β-catenin signaling. Proc Natl Acad Sci U S A 2021; 118:e2020100118. [PMID: 34172578 PMCID: PMC8256036 DOI: 10.1073/pnas.2020100118] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Job syndrome is a rare genetic disorder caused by STAT3 mutations and primarily characterized by immune dysfunction along with comorbid skeleton developmental abnormalities including osteopenia, recurrent fracture of long bones, and scoliosis. So far, there is no definitive cure for the skeletal defects in Job syndrome, and treatments are limited to management of clinical symptoms only. Here, we have investigated the molecular mechanism whereby Stat3 regulates skeletal development and osteoblast differentiation. We showed that removing Stat3 function in the developing limb mesenchyme or osteoprogenitor cells in mice resulted in shortened and bow limbs with multiple fractures in long bones that resembled the skeleton symptoms in the Job Syndrome. However, Stat3 loss did not alter chondrocyte differentiation and hypertrophy in embryonic development, while osteoblast differentiation was severely reduced. Genome-wide transcriptome analyses as well as biochemical and histological studies showed that Stat3 loss resulted in down-regulation of Wnt/β-catenin signaling. Restoration of Wnt/β-catenin signaling by injecting BIO, a small molecule inhibitor of GSK3, or crossing with a Lrp5 gain of function (GOF) allele, rescued the bone reduction phenotypes due to Stat3 loss to a great extent. These studies uncover the essential functions of Stat3 in maintaining Wnt/β-catenin signaling in early mesenchymal or osteoprogenitor cells and provide evidence that bone defects in the Job Syndrome are likely caused by Wnt/β-catenin signaling reduction due to reduced STAT3 activities in bone development. Enhancing Wnt/β-catenin signaling could be a therapeutic approach to reduce bone symptoms of Job syndrome patients.
Collapse
Affiliation(s)
- Prem Swaroop Yadav
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115
| | - Shuhao Feng
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115
| | - Qian Cong
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115
| | - Hanjun Kim
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115
| | - Yuchen Liu
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115
| | - Yingzi Yang
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115;
- Harvard Stem Cell Institute, Cambridge, MA 02138
| |
Collapse
|
195
|
Wang H, Wei P, Zhang Y, Li Y, Yin L. LncRNA TCONS_00023297 Regulates the Balance of Osteogenic and Adipogenic Differentiation in Bone Marrow Mesenchymal Stem Cells and the Coupling Process of Osteogenesis and Angiogenesis. Front Cell Dev Biol 2021; 9:697858. [PMID: 34262909 PMCID: PMC8274487 DOI: 10.3389/fcell.2021.697858] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 05/27/2021] [Indexed: 12/01/2022] Open
Abstract
Long noncoding RNA (lncRNA) is a noncoding RNA with a length of more than 200 bases. It plays an important role in the occurrence and development of diseases. Research on lncRNAs has received increasing attention. Bone is an important organ of the human body. As the population ages, the incidence of osteoporosis gradually increases. The mechanism of action of lncRNAs in the development of osteoporosis is unclear. The imbalance between osteogenic and adipogenic differentiation in bone marrow mesenchymal stem cells (hBMSCs) and the coupling process of osteogenesis and angiogenesis plays an important role in the development of osteoporosis. Therefore, this study focused on the mechanism by which lncRNAs regulate the osteogenic differentiation of bone marrow mesenchymal stem cells and the mechanism of action of lncRNAs in bone metabolism. The expression of lncRNAs in the osteogenic differentiation of hBMSCs was detected by lncRNA microarray. Real-time quantitative PCR was used to detect the expression changes of lncRNA and osteogenic genes during hBMSC osteogenic and adipogenic differentiation. The ceRNA mechanisms were detected by RIP and luciferase reporter gene assays. The effect of lncRNAs on the osteogenesis–angiogenesis coupling process was detected by Transwell assays. TCONS_00023297 increased expression during osteogenic differentiation; TCONS_00023297 overexpression promoted osteogenic differentiation of hBMSCs; BMP2 regulated TCONS_00023297 expression in a concentration- and time-dependent manner; TCONS_00023297 regulated miR-608 via a ceRNA mechanism; TCONS_00023297 inhibited hBMSC adipogenic differentiation; and TCONS_00023297 promoted VEGF secretion by hBMSCs. TCONS_00023297 regulates osteogenic differentiation, adipogenic differentiation, and osteogenic–angiogenic coupling of hBMSCs via the TCONS_00023297/miR-608/RUNX2/SHH signaling axis.
Collapse
Affiliation(s)
- Haitao Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Peng Wei
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuebai Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Li Yin
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
196
|
Genistein inhibits chondrogenic differentiation and mineralization of ATDC5 cells. Biochem Biophys Res Commun 2021; 566:123-128. [PMID: 34119824 DOI: 10.1016/j.bbrc.2021.05.101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 05/31/2021] [Indexed: 01/07/2023]
Abstract
Isoflavones are phytoestrogens abundant in leguminous crops and are used to prevent a variety of hormonal disorders. In the present study, the effects of genistein and daidzein on the chondrogenic differentiation of ATDC5 cells were investigated. Genistein (10 μM) treatment markedly reduced production of sulfated proteoglycans and collagen fibers in the ATDC5 cells. Genistein suppressed the expression of genes involved in chondrocyte differentiation such as Sox9, Col2a1, Col10a1, Acan, and Tgfb1. Additionally, genistein significantly decreased calcium deposition in ATDC5 cells during chondrogenic differentiation; however, it increased calcification under non-chondrogenic mineralizing conditions. Daidzein exhibited a similar effect of suppressing chondrogenesis in ATDC5 cells, although its efficacy was 10-times lower than that of genistein. These findings suggest that a high concentration of genistein inhibits chondrogenesis and chondrogenic mineralization, whereas it enhances non-chondrogenic mineralization.
Collapse
|
197
|
It is time to crowd your cell culture media - Physicochemical considerations with biological consequences. Biomaterials 2021; 275:120943. [PMID: 34139505 DOI: 10.1016/j.biomaterials.2021.120943] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 05/24/2021] [Accepted: 05/29/2021] [Indexed: 12/12/2022]
Abstract
In vivo, the interior and exterior of cells is populated by various macromolecules that create an extremely crowded milieu. Yet again, in vitro eukaryotic cell culture is conducted in dilute culture media that hardly imitate the native tissue density. Herein, the concept of macromolecular crowding is discussed in both intracellular and extracellular context. Particular emphasis is given on how the physicochemical properties of the crowding molecules govern and determine kinetics, equilibria and mechanism of action of biochemical and biological reactions, processes and functions. It is evidenced that we are still at the beginning of appreciating, let alone effectively implementing, the potential of macromolecular crowding in permanently differentiated and stem cell culture systems.
Collapse
|
198
|
Dalle Carbonare L, Bertacco J, Marchetto G, Cheri S, Deiana M, Minoia A, Tiso N, Mottes M, Valenti MT. Methylsulfonylmethane enhances MSC chondrogenic commitment and promotes pre-osteoblasts formation. Stem Cell Res Ther 2021; 12:326. [PMID: 34090529 PMCID: PMC8180127 DOI: 10.1186/s13287-021-02396-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 05/18/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Methylsulfonylmethane (MSM) is a nutraceutical compound which has been indicated to counteract osteoarthritis, a cartilage degenerative disorder. In addition, MSM has also been shown to increase osteoblast differentiation. So far, few studies have investigated MSM role in the differentiation of mesenchymal stem cells (MSCs), and no study has been performed to evaluate its overall effects on both osteogenic and chondrogenic differentiation. These two mutually regulated processes share the same progenitor cells. METHODS Therefore, with the aim to evaluate the effects of MSM on chondrogenesis and osteogenesis, we analyzed the expression of SOX9, RUNX2, and SP7 transcription factors in vitro (mesenchymal stem cells and chondrocytes cell lines) and in vivo (zebrafish model). Real-time PCR as well Western blotting, immunofluorescence, and specific in vitro and in vivo staining have been performed. Student's paired t test was used to compare the variation between the groups. RESULTS Our data demonstrated that MSM modulates the expression of differentiation-related genes both in vitro and in vivo. The increased SOX9 expression suggests that MSM promotes chondrogenesis in treated samples. In addition, RUNX2 expression was not particularly affected by MSM while SP7 expression increased in all MSM samples/model analyzed. As SP7 is required for the final commitment of progenitors to preosteoblasts, our data suggest a role of MSM in promoting preosteoblast formation. In addition, we observed a reduced expression of the osteoclast-surface receptor RANK in larvae and in scales as well as a reduced pERK/ERK ratio in fin and scale of MSM treated zebrafish. CONCLUSIONS In conclusion, our study provides new insights into MSM mode of action and suggests that MSM is a useful tool to counteract skeletal degenerative diseases by targeting MSC commitment and differentiation.
Collapse
Affiliation(s)
- Luca Dalle Carbonare
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Jessica Bertacco
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie, 10, 37100, Verona, Italy
| | - Giulia Marchetto
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Samuele Cheri
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Michela Deiana
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Arianna Minoia
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Natascia Tiso
- Department of Biology, University of Padova, I-35131, Padova, Italy
| | - Monica Mottes
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie, 10, 37100, Verona, Italy
| | - Maria Teresa Valenti
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy.
| |
Collapse
|
199
|
Schott NG, Friend NE, Stegemann JP. Coupling Osteogenesis and Vasculogenesis in Engineered Orthopedic Tissues. TISSUE ENGINEERING. PART B, REVIEWS 2021; 27:199-214. [PMID: 32854589 PMCID: PMC8349721 DOI: 10.1089/ten.teb.2020.0132] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/25/2020] [Indexed: 12/20/2022]
Abstract
Inadequate vascularization of engineered tissue constructs is a main challenge in developing a clinically impactful therapy for large, complex, and recalcitrant bone defects. It is well established that bone and blood vessels form concomitantly during development, as well as during repair after injury. Endothelial cells (ECs) and mesenchymal stromal cells (MSCs) are known to be key players in orthopedic tissue regeneration and vascularization, and these cell types have been used widely in tissue engineering strategies to create vascularized bone. Coculture studies have demonstrated that there is crosstalk between ECs and MSCs that can lead to synergistic effects on tissue regeneration. At the same time, the complexity in fabricating, culturing, and characterizing engineered tissue constructs containing multiple cell types presents a challenge in creating multifunctional tissues. In particular, the timing, spatial distribution, and cell phenotypes that are most conducive to promoting concurrent bone and vessel formation are not well understood. This review describes the processes of bone and vascular development, and how these have been harnessed in tissue engineering strategies to create vascularized bone. There is an emphasis on interactions between ECs and MSCs, and the culture systems that can be used to understand and control these interactions within a single engineered construct. Developmental engineering strategies to mimic endochondral ossification are discussed as a means of generating vascularized orthopedic tissues. The field of tissue engineering has made impressive progress in creating tissue replacements. However, the development of larger, more complex, and multifunctional engineered orthopedic tissues will require a better understanding of how osteogenesis and vasculogenesis are coupled in tissue regeneration. Impact statement Vascularization of large engineered tissue volumes remains a challenge in developing new and more biologically functional bone grafts. A better understanding of how blood vessels develop during bone formation and regeneration is needed. This knowledge can then be applied to develop new strategies for promoting both osteogenesis and vasculogenesis during the creation of engineered orthopedic tissues. This article summarizes the processes of bone and blood vessel development, with a focus on how endothelial cells and mesenchymal stromal cells interact to form vascularized bone both during development and growth, as well as tissue healing. It is meant as a resource for tissue engineers who are interested in creating vascularized tissue, and in particular to those developing cell-based therapies for large, complex, and recalcitrant bone defects.
Collapse
Affiliation(s)
- Nicholas G. Schott
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Nicole E. Friend
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Jan P. Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
200
|
Sawadkar P, Mandakhbayar N, Patel KD, Buitrago JO, Kim TH, Rajasekar P, Lali F, Kyriakidis C, Rahmani B, Mohanakrishnan J, Dua R, Greco K, Lee JH, Kim HW, Knowles J, García-Gareta E. Three dimensional porous scaffolds derived from collagen, elastin and fibrin proteins orchestrate adipose tissue regeneration. J Tissue Eng 2021; 12:20417314211019238. [PMID: 34104389 PMCID: PMC8165536 DOI: 10.1177/20417314211019238] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/04/2021] [Indexed: 12/18/2022] Open
Abstract
Current gold standard to treat soft tissue injuries caused by trauma and pathological condition are autografts and off the shelf fillers, but they have inherent weaknesses like donor site morbidity, immuno-compatibility and graft failure. To overcome these limitations, tissue-engineered polymers are seeded with stem cells to improve the potential to restore tissue function. However, their interaction with native tissue is poorly understood so far. To study these interactions and improve outcomes, we have fabricated scaffolds from natural polymers (collagen, fibrin and elastin) by custom-designed processes and their material properties such as surface morphology, swelling, wettability and chemical cross-linking ability were characterised. By using 3D scaffolds, we comprehensive assessed survival, proliferation and phenotype of adipose-derived stem cells in vitro. In vivo, scaffolds were seeded with adipose-derived stem cells and implanted in a rodent model, with X-ray microtomography, histology and immunohistochemistry as read-outs. Collagen-based materials showed higher cell adhesion and proliferation in vitro as well as higher adipogenic properties in vivo. In contrast, fibrin demonstrated poor cellular and adipogenesis properties but higher angiogenesis. Elastin formed the most porous scaffold, with cells displaying a non-aggregated morphology in vitro while in vivo elastin was the most degraded scaffold. These findings of how polymers present in the natural polymers mimicking ECM and seeded with stem cells affect adipogenesis in vitro and in vivo can open avenues to design 3D grafts for soft tissue repair.
Collapse
Affiliation(s)
- Prasad Sawadkar
- Regenerative Biomaterials Group, The RAFT Institute and The Griffin Institute, Northwick Park & Saint Mark's Hospital, London, UK.,Division of Surgery and Interventional Science, University College London, London, UK.,UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea
| | - Nandin Mandakhbayar
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science & BK21 Plus NBM Global Research Centre for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea.,Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, Republic of Korea
| | - Kapil D Patel
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea.,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science & BK21 Plus NBM Global Research Centre for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea.,Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, London, UK
| | - Jennifer Olmas Buitrago
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea.,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science & BK21 Plus NBM Global Research Centre for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea
| | - Tae Hyun Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science & BK21 Plus NBM Global Research Centre for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea.,R&D Center, TE Bios Co, Osong, Republic of Korea
| | - Poojitha Rajasekar
- Division of Respiratory Medicine, University of Nottingham, Nottingham, UK
| | - Ferdinand Lali
- Division of Surgery and Interventional Science, University College London, London, UK.,The Griffin Institute, Northwick Park and St Mark's Hospital, London, UK
| | - Christos Kyriakidis
- Regenerative Biomaterials Group, The RAFT Institute and The Griffin Institute, Northwick Park & Saint Mark's Hospital, London, UK
| | - Benyamin Rahmani
- Department of Mechanical Engineering, University College London, London, UK
| | - Jeviya Mohanakrishnan
- Regenerative Biomaterials Group, The RAFT Institute and The Griffin Institute, Northwick Park & Saint Mark's Hospital, London, UK
| | - Rishbha Dua
- Regenerative Biomaterials Group, The RAFT Institute and The Griffin Institute, Northwick Park & Saint Mark's Hospital, London, UK
| | - Karin Greco
- Division of Surgery and Interventional Science, University College London, London, UK.,The Griffin Institute, Northwick Park and St Mark's Hospital, London, UK
| | - Jung-Hwan Lee
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea.,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science & BK21 Plus NBM Global Research Centre for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea.,Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, Republic of Korea
| | - Hae-Won Kim
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea.,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science & BK21 Plus NBM Global Research Centre for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea.,Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, Republic of Korea
| | - Jonathan Knowles
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea.,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science & BK21 Plus NBM Global Research Centre for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea.,Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, Republic of Korea.,Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, London, UK
| | - Elena García-Gareta
- Regenerative Biomaterials Group, The RAFT Institute and The Griffin Institute, Northwick Park & Saint Mark's Hospital, London, UK.,Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, London, UK
| |
Collapse
|