1
|
Fan W, Liu Y, Hou F, Zhao F, Wu B, Jiang W. In vitro/ in vivo evaluation of double crosslinked bone glue with different degrees. Biotechnol Genet Eng Rev 2024; 40:3047-3063. [PMID: 37078415 DOI: 10.1080/02648725.2023.2203008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/11/2023] [Indexed: 04/21/2023]
Abstract
Successful bone fragment fixation is a crucial factor in bone fracture healing, the fixation of crushed bone fragments could hinder bone fracture healing. Thus, ideal bone glues to effectively adhere and splice comminuted bone fragments are needed in clinical. Herein, an osteoinductive and biodegradable double cross-linked bone glue (GelMA-oDex-AMBGN) was constructed through Schiff's base reaction between commercial GelMA (with different substitution degrees of amino groups) and Odex mixed with amine-modified mesoporous bioactive glass nanoparticles (AMBGN), followed by crosslink with blue light irradiation. The GelMA-oDex-AMBGN bone glue successfully adhered and spliced the comminuted bone fragments of isolated rat skulls. GelMA-oDex-AMBGN promoted the proliferation of 3T3 cells and enhanced the expression of osteogenic proteins Runx2 and OCN in vitro. In rat cranial critical-sized defect models, GelMA-oDex-AMBGNs with different substitution degrees significantly increased the new bone contents at the fracture defect sites and promoted bone tissue regeneration in vivo. In conclusion, the double cross-linked bone glue (GelMA-oDex-AMBGN) was successfully constructed and can induce bone regeneration. Additionally, there was no significant difference in osteogenic activity among GelMA-oDex-AMBGNs with different substitution degrees and the equal content of AMBGN.
Collapse
Affiliation(s)
- Wei Fan
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yijie Liu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Fushan Hou
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Feng Zhao
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Binqiang Wu
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Weimin Jiang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
2
|
Ohyama Y, Ohta Y, Sugama R, Minoda Y, Masuda S, Terai H, Nakamura H. Effect of recombinant irisin on recombinant human bone morphogenetic protein-2 induced osteogenesis and osteoblast differentiation. Biochem Biophys Res Commun 2024; 734:150787. [PMID: 39368373 DOI: 10.1016/j.bbrc.2024.150787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 10/01/2024] [Indexed: 10/07/2024]
Abstract
Osteoporotic fragility fractures substantially impact aging societies, necessitating long-term care and increasing healthcare costs. Myokine irisin, secreted by skeletal muscle, influences bone metabolism; however, a comprehensive understanding of the mechanisms by which irisin affects bone metabolism is still lacking. Therefore, this study aimed to explore the effects of irisin on osteogenesis and osteoblast differentiation triggered by bone morphogenetic protein-2 (BMP-2). We used 4-week-old male ICR mice and implanted polyethylene glycol pellets containing recombinant human BMP-2 (rh-BMP-2) into the left dorsal muscle pouch. Mice received weekly intraperitoneal injections of either phosphate-buffered saline or recombinant irisin (re-irisin). Ectopic bone formation was evaluated 3 weeks post-surgery using micro-computed tomography (μ-CT) and histological analysis. In vitro experiments, C2C12 cells were treated with or without rh-BMP-2 and re-irisin, and we assessed osteoblast differentiation markers, e.g., runt-related transcription factor 2, alkaline phosphatase, osteocalcin, and osteopontin, using real-time reverse transcription-polymerase chain reaction. The μ-CT analyses showed that re-irisin significantly increased bone mineral content and bone volume of ectopic bones newly formed by rh-BMP-2. The gene expressions of the osteoblast markers were significantly increased by rh-BMP-2 and further upregulated by re-irisin. The treatment of cyclic AMP response element-binding protein (CREB) small interfering RNA attenuated these effects, suggesting that CREB signaling pathway was involved in rh-BMP-2/re-irisin-induced osteoblastic differentiation. This study demonstrates the potential of irisin to enhance osteogenesis through BMP signaling, offering insights for osteoporosis treatment and highlighting irisin as a promising therapeutic target for improving bone health and extending a healthy lifespan.
Collapse
Affiliation(s)
- Yohei Ohyama
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Japan; Department of Orthopaedic Surgery, Osaka Metropolitan University Graduate School of Medicine, Japan
| | - Yoichi Ohta
- Department of Orthopaedic Surgery, Osaka Metropolitan University Graduate School of Medicine, Japan.
| | - Ryo Sugama
- Department of Orthopaedic Surgery, Osaka Metropolitan University Graduate School of Medicine, Japan
| | - Yukihide Minoda
- Department of Orthopaedic Surgery, Osaka Metropolitan University Graduate School of Medicine, Japan
| | - Sho Masuda
- Department of Orthopaedic Surgery, Osaka Metropolitan University Graduate School of Medicine, Japan
| | - Hidetomi Terai
- Department of Orthopaedic Surgery, Osaka Metropolitan University Graduate School of Medicine, Japan
| | - Hiroaki Nakamura
- Department of Orthopaedic Surgery, Osaka Metropolitan University Graduate School of Medicine, Japan
| |
Collapse
|
3
|
Wagner J, Bayer L, Loger K, Acil Y, Kurz S, Spille J, Ahlhelm M, Ingwersen LC, Jonitz-Heincke A, Sedaghat S, Wiltfang J, Naujokat H. In vivo endocultivation of CAD/CAM hybrid scaffolds in the omentum majus in miniature pigs. J Craniomaxillofac Surg 2024; 52:1259-1266. [PMID: 39198129 DOI: 10.1016/j.jcms.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/27/2024] [Indexed: 09/01/2024] Open
Abstract
PURPOSE Correction of bony mandibular defects is a challenge in oral and maxillofacial surgery due to aesthetic and functional requirements. This study investigated the potential of a novel hybrid scaffold for bone regeneration and degradation assessment of the ceramic within the omentum majus over 6 months and the extent to which rhBMP-2 as a growth factor, alone or combined with a hydrogel, affects regeneration. MATERIALS AND METHODS In this animal study, 10 Göttingen minipigs each had one scaffold implanted in the greater omentum. Five animals had scaffolds loaded with a collagen hydrogel and rhBMP-2, and the other five animals (control group) had scaffolds loaded with rhBMP-2 only. Fluorochrome injections and computed tomography (CT) were performed regularly. After 6 months, the animals were euthanized, and samples were collected for microCT and histological evaluations. RESULTS Fluorescent and light microscopic and a CT morphological density evaluation showed continuous bone growth until week 16 in both groups. Regarding the ratio of bone attachment to the Zr02 support struts, the rhBMP-2 loaded collagen hydrogel group showed with 63% a significantly higher attachment (p > 0.001) than the rhBMP-2 control group (49%). CONCLUSION In this study, bone growth was induced in all omentum majus specimens until post-operative week 16. Furthermore, hydrogel and rhBMP-2 together resulted in better bone-scaffold integration than rhBMP-2 alone. Further studies should investigate whether implantation of the scaffolds in the jaw after an appropriate period of bone regeneration leads to a stable situation and the desired results.
Collapse
Affiliation(s)
- Juliane Wagner
- Department of Oral and Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany; Cluster of Excellence, Precision Medicine in Inflammation, Christian-Albrechts-University of Kiel, Kiel, Germany.
| | - Lennart Bayer
- Department of Oral and Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Klaas Loger
- Department of Oral and Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Yahya Acil
- Department of Oral and Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Sascha Kurz
- ZESBO - Center for Research on Musculoskeletal Systems, Leipzig University, Leipzig, Germany
| | - Johannes Spille
- Department of Oral and Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Matthias Ahlhelm
- Fraunhofer Institute for Ceramic Technologies and Systems, IKTS, Dresden, Germany
| | - Lena-Christin Ingwersen
- Biomechanics and Implant Technology Research Laboratory, Department of Orthopaedics, Rostock University Medical Center, Rostock, Germany
| | - Anika Jonitz-Heincke
- Biomechanics and Implant Technology Research Laboratory, Department of Orthopaedics, Rostock University Medical Center, Rostock, Germany
| | - Sam Sedaghat
- Department of Diagnostic and Interventional Radiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Jörg Wiltfang
- Department of Oral and Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Hendrik Naujokat
- Department of Oral and Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
4
|
Doyle SE, Cazzola CN, Coleman CM. Design considerations when creating a high throughput screen-compatible in vitro model of osteogenesis. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100184. [PMID: 39313131 DOI: 10.1016/j.slasd.2024.100184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/06/2024] [Accepted: 09/20/2024] [Indexed: 09/25/2024]
Abstract
Inducing osteogenic differentiation in vitro is useful for the identification and development of bone regeneration therapies as well as modelling bone disorders. To couple in vitro models with high throughput screening techniques retains the assay's relevance in research while increasing its therapeutic impact. Miniaturizing, automating and/or digitalizing in vitro assays will reduce the required quantity of cells, biologic stimulants, culture/output assay reagents, time and cost. This review highlights the design and workflow considerations for creating a high throughput screen-compatible model of osteogenesis, comparing and contrasting osteogenic cell type, assay fabrication and culture methodology, osteogenic induction approach and repurposing existing output techniques.
Collapse
Affiliation(s)
- Stephanie E Doyle
- Regenerative Medicine Institute, School of Medicine, College of Medicine, Nursing and Health Science, University of Galway, Galway City, County Galway H91 FD82, Ireland.
| | - Courtney N Cazzola
- Regenerative Medicine Institute, School of Medicine, College of Medicine, Nursing and Health Science, University of Galway, Galway City, County Galway H91 FD82, Ireland
| | - Cynthia M Coleman
- Regenerative Medicine Institute, School of Medicine, College of Medicine, Nursing and Health Science, University of Galway, Galway City, County Galway H91 FD82, Ireland
| |
Collapse
|
5
|
Wei S, Wu Q, Cao C, Yang Z, Shi J, Huang J, He H, Lai Y, Li J. A mechanism of action-reflective, dual cell-based bioassay for determining the bioactivity of sclerostin-neutralizing antibodies. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100187. [PMID: 39389544 DOI: 10.1016/j.slasd.2024.100187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
Osteoporosis is a major threat to the elderly worldwide. The Wnt signaling pathway plays a critical role in bone development and homeostasis. Sclerostin, a Wnt ligand inhibitor, competes with Wnt ligands for low-density lipoprotein receptor-related protein 5 or 6 (LRP5/6) on osteoblasts, thereby suppressing bone formation. Sclerostin-neutralizing monoclonal antibodies (mAbs) have emerged as a potential bone-forming therapy for osteoporosis. A cell-based bioassay which determines the relative activity of a product, related to its mechanism of action, is of great importance from drug discovery to quality control and batch release. Currently used cell-based bioassays for sclerostin-neutralizing mAbs usually use Wnt1 or Wnt3a to stimulate the Wnt pathway; sclerostin is a direct inhibitor of Wnt1 but not Wnt3a. Wnt1 is a highly hydrophobic protein that binds to the producing cell membrane and acts in a juxtacrine manner to stimulate the Wnt pathway in neighboring cells. Bioassays for drugs that induce Wnt1 signaling should be performed in a juxtacrine manner. Here, we present a mechanism of action-reflective, dual cell-based reporter gene assay. In this assay, Wnt1 producer cells are co-cultured with cells containing the Wnt reporter genes, Wnt1 on the producer cells activates the Wnt signaling pathway in the reporter cells that are in direct cell-to-cell contact, and sclerostin-neutralizing mAbs specifically and effectively antagonize the sclerostin-mediated Wnt reporter gene suppression. This bioassay demonstrates good specificity, accuracy, linearity, and precision and is suitable for quality control, stability testing, batch release, and biosimilarity assessment of sclerostin-neutralizing mAbs.
Collapse
Affiliation(s)
- Suzhen Wei
- Zhuhai United Biopharma Co., Ltd, 399 Airport West Road, Zhuhai, Guangdong, China
| | - Qiang Wu
- Zhuhai United Laboratories Co., Ltd, 2428 Anji Road, Zhuhai, Guangdong, China
| | - Chunlai Cao
- Zhuhai United Biopharma Co., Ltd, 399 Airport West Road, Zhuhai, Guangdong, China
| | - Zhuoni Yang
- Zhuhai United Biopharma Co., Ltd, 399 Airport West Road, Zhuhai, Guangdong, China
| | - Jianrui Shi
- Zhuhai United Biopharma Co., Ltd, 399 Airport West Road, Zhuhai, Guangdong, China
| | - Jingqun Huang
- Zhuhai United Biopharma Co., Ltd, 399 Airport West Road, Zhuhai, Guangdong, China
| | - Hua He
- Zhuhai United Biopharma Co., Ltd, 399 Airport West Road, Zhuhai, Guangdong, China
| | - Yongjie Lai
- Department of Microbiology and Immunology, Zunyi Medical University (Zhuhai Campus), 368 Golden Coast Avenue, Zhuhai, Guangdong, China.
| | - Jing Li
- Zhuhai United Biopharma Co., Ltd, 399 Airport West Road, Zhuhai, Guangdong, China; Zhuhai United Laboratories Co., Ltd, 2428 Anji Road, Zhuhai, Guangdong, China.
| |
Collapse
|
6
|
Toita R, Shimizu Y, Shimizu E, Deguchi T, Tsuchiya A, Kang JH, Kitamura M, Kato A, Yamada H, Yamaguchi S, Kasahara S. Collagen patches releasing phosphatidylserine liposomes guide M1-to-M2 macrophage polarization and accelerate simultaneous bone and muscle healing. Acta Biomater 2024; 187:51-65. [PMID: 39159714 DOI: 10.1016/j.actbio.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/22/2024] [Accepted: 08/11/2024] [Indexed: 08/21/2024]
Abstract
Bilateral communication between bones and muscles is essential for healing composite bone-muscle injuries from orthopedic surgeries and trauma. However, these injuries are often characterized by exaggerated inflammation, which can disrupt bone-muscle crosstalk, thereby seriously delaying the healing of either tissue. Existing approaches are largely effective at healing single tissues. However, simultaneous healing of multiple tissues remains challenging, with little research conducted to date. Here we introduce collagen patches that overcome this overlooked issue by harnessing the plasticity of macrophage phenotypes. Phosphatidylserine liposomes (PSLs) capable of shifting the macrophage phenotype from inflammatory M1 into anti-inflammatory/prohealing M2 were coated on collagen patches via a layer-by-layer method. Original collagen patches failed to improve tissue healing under inflammatory conditions coordinated by M1 macrophages. In contrast, PSL-coated collagen patches succeeded in accelerating bone and muscle healing by inducing a microenvironment dominated by M2 macrophages. In cell experiments, differentiation of preosteoblasts and myoblasts was completely inhibited by secretions of M1 macrophages but unaffected by those of M2 macrophages. RNA-seq analysis revealed that type I interferon and interleukin-6 signaling pathways were commonly upregulated in preosteoblasts and myoblasts upon stimulation with M1 macrophage secretions, thereby compromising their differentiation. This study demonstrates the benefit of PSL-mediated M1-to-M2 macrophage polarization for simultaneous bone and muscle healing, offering a potential strategy toward simultaneous regeneration of multiple tissues. STATEMENT OF SIGNIFICANCE: Existing approaches for tissue regeneration, which primarily utilize growth factors, have been largely effective at healing single tissues. However, simultaneous healing of multiple tissues remains challenging and has been little studied. Here we demonstrate that collagen patches releasing phosphatidylserine liposomes (PSLs) promote M1-to-M2 macrophage polarization and are effective for simultaneous healing of bone and muscle. Transcriptome analysis using next-generation sequencing reveals that differentiation of preosteoblasts and myoblasts is inhibited by the secretions of M1 macrophages but promoted by those of M2 macrophages, highlighting the importance of timely regulation of M1-to-M2 polarization in tissue regeneration. These findings provide new insight to tissue healing of multiple tissues.
Collapse
Affiliation(s)
- Riki Toita
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka, 563-8577, Japan; AIST-Osaka University Advanced Photonics and Biosensing Open Innovation Laboratory, AIST, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Yuki Shimizu
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka, 563-8577, Japan
| | - Eiko Shimizu
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka, 563-8577, Japan
| | - Tomonori Deguchi
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka, 563-8577, Japan
| | - Akira Tsuchiya
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Jeong-Hun Kang
- National Cerebral and Cardiovascular Center Research Institute, 6-1 Shinmachi, Kishibe, Suita, Osaka, 564-8565, Japan
| | - Masahiro Kitamura
- NGK Spark Plug-AIST Healthcare Materials Cooperative Research Laboratory, Anagahora, Shimo-shidami, Moriyama-ku, Nagoya, Aichi 463-8560, Japan; Niterra Co., Ltd., 2808 Iwasaki, Komaki, Aichi 485-8510, Japan
| | - Atsushi Kato
- NGK Spark Plug-AIST Healthcare Materials Cooperative Research Laboratory, Anagahora, Shimo-shidami, Moriyama-ku, Nagoya, Aichi 463-8560, Japan; Niterra Co., Ltd., 2808 Iwasaki, Komaki, Aichi 485-8510, Japan
| | - Hideto Yamada
- Niterra Co., Ltd., 2808 Iwasaki, Komaki, Aichi 485-8510, Japan
| | - Shogo Yamaguchi
- Niterra Co., Ltd., 2808 Iwasaki, Komaki, Aichi 485-8510, Japan
| | | |
Collapse
|
7
|
Nakano N, Tashiro E, Shimada T, Ebisawa M, Kojima S, Ayabe K, Yamamoto Y, Maeda S, Itoh F, Itoh S. Involvement of mitogen- and stress-activated protein kinase 1 in BMP-6-induced chondrocyte differentiation. J Biol Chem 2024; 300:107806. [PMID: 39307301 PMCID: PMC11541777 DOI: 10.1016/j.jbc.2024.107806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/27/2024] [Accepted: 09/13/2024] [Indexed: 10/27/2024] Open
Abstract
Bone morphogenetic proteins (BMPs) are involved in several cellular responsive actions, such as development, cell differentiation, and apoptosis, via their specific transmembrane receptors. In particular, BMPs promote the differentiation and maturation of bone and cartilage from mesenchymal stem cells. Based on comprehensive analyses performed with a large number of antibodies, mitogen- and stress-activated protein kinase (MSK)1 was found to be immediately phosphorylated in the mouse chondrocyte precursor cell line, ATDC5, upon BMP-6 stimulation. The overexpression and knockdown of MSK1 in ATDC5 cells also enhanced and suppressed BMP-6-induced chondrocyte differentiation, respectively. Similar to ATDC5 cells, an ex vivo organ culture system using mouse embryonic metatarsal bones also demonstrated that BMP-6-mediated MSK1 activation might play a role in chondrocyte differentiation. Using several inhibitors, the p38 kinase pathway was confirmed to be implicated in BMP-6-induced phosphorylation of MSK1. Furthermore, MSK1 mutants lacking kinase activities and those lacking serine/threonine residues targeted by p38 kinase severely impaired their ability to potentiate BMP-6-induced chondrogenic differentiation of ATDC5 cells. Interestingly, a loss-of-function study for Smad4 perturbed BMP-6-induced phosphorylation of p38 kinase to inhibit BMP-6-mediated chondrocyte differentiation via MSK1 activation. Overall, both Smad-dependent and independent pathways require BMP-6-induced chondrocyte differentiation via MSK1 activation in ATDC5 cells.
Collapse
Affiliation(s)
- Naoko Nakano
- Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo, Japan
| | - Etsu Tashiro
- Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo, Japan
| | - Takayuki Shimada
- Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo, Japan
| | - Masayasu Ebisawa
- Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo, Japan
| | - Sayaka Kojima
- Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo, Japan
| | - Kaho Ayabe
- Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo, Japan
| | - Yohei Yamamoto
- Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo, Japan
| | - Shingo Maeda
- Department of Bone and Joint Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Kagoshima, Japan
| | - Fumiko Itoh
- Laboratory of Stem Cell Regulation, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Susumu Itoh
- Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo, Japan.
| |
Collapse
|
8
|
Yu Y, Chen R, Chen X, Wang J, Liu C. Regulating the bioactivity of non-glycosylated recombinant human bone morphogenetic protein-2 to enhance bone regeneration. Bioact Mater 2024; 38:169-180. [PMID: 38711759 PMCID: PMC11070760 DOI: 10.1016/j.bioactmat.2024.04.018] [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: 11/29/2023] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 05/08/2024] Open
Abstract
Recombinant human bone morphogenetic protein-2 (rhBMP-2) is the predominant growth factor that effectively induces osteogenic differentiation in orthopedic procedures. However, the bioactivity and stability of rhBMP-2 are intrinsically associated with its sequence, structure, and storage conditions. In this study, we successfully determined the amino acid sequence and protein secondary structure model of non-glycosylated rhBMP-2 expressed by an E. coli expression system through X-ray crystal structure analysis. Furthermore, we observed that acidic storage conditions enhanced the proliferative and osteoinductive activity of rhBMP-2. Although the osteogenic activity of non-glycosylated rhBMP-2 is relatively weaker compared to glycosylated rhBMP-2; however, this discrepancy can be mitigated by incorporating exogenous chaperone molecules. Overall, such information is crucial for rationalizing the design of stabilization methods and enhancing the bioactivity of rhBMP-2, which may also be applicable to other growth factors.
Collapse
Affiliation(s)
- Yuanman Yu
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Rui Chen
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Xinye Chen
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Jing Wang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, PR China
| |
Collapse
|
9
|
Rich J, Bennaroch M, Notel L, Patalakh P, Alberola J, Issa F, Opolon P, Bawa O, Rondof W, Marchais A, Dessen P, Meurice G, Le-Gall M, Polrot M, Ser-Le Roux K, Mamchaoui K, Droin N, Raslova H, Maire P, Geoerger B, Pirozhkova I. DiPRO1 distinctly reprograms muscle and mesenchymal cancer cells. EMBO Mol Med 2024; 16:1840-1885. [PMID: 39009887 PMCID: PMC11319797 DOI: 10.1038/s44321-024-00097-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 07/17/2024] Open
Abstract
We have recently identified the uncharacterized ZNF555 protein as a component of a productive complex involved in the morbid function of the 4qA locus in facioscapulohumeral dystrophy. Subsequently named DiPRO1 (Death, Differentiation, and PROliferation related PROtein 1), our study provides substantial evidence of its role in the differentiation and proliferation of human myoblasts. DiPRO1 operates through the regulatory binding regions of SIX1, a master regulator of myogenesis. Its relevance extends to mesenchymal tumors, such as rhabdomyosarcoma (RMS) and Ewing sarcoma, where DiPRO1 acts as a repressor via the epigenetic regulators TIF1B and UHRF1, maintaining methylation of cis-regulatory elements and gene promoters. Loss of DiPRO1 mimics the host defense response to virus, awakening retrotransposable repeats and the ZNF/KZFP gene family. This enables the eradication of cancer cells, reprogramming the cellular decision balance towards inflammation and/or apoptosis by controlling TNF-α via NF-kappaB signaling. Finally, our results highlight the vulnerability of mesenchymal cancer tumors to si/shDiPRO1-based nanomedicines, positioning DiPRO1 as a potential therapeutic target.
Collapse
Affiliation(s)
- Jeremy Rich
- UMR8126 CNRS, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Melanie Bennaroch
- UMR8126 CNRS, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Laura Notel
- UMR8126 CNRS, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Polina Patalakh
- UMR8126 CNRS, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Julien Alberola
- UMR8126 CNRS, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Fayez Issa
- INSERM U1016, CNRS UMR 8104, Institut Cochin, Université Paris-Cité, Paris, France
| | - Paule Opolon
- Pathology and Cytology Section, UMS AMMICA, CNRS, INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Olivia Bawa
- Pathology and Cytology Section, UMS AMMICA, CNRS, INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Windy Rondof
- Bioinformatics Platform, UMS AMMICA, CNRS, INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer campus, INSERM U1015, Université Paris-Saclay, Villejuif, France
| | - Antonin Marchais
- Bioinformatics Platform, UMS AMMICA, CNRS, INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer campus, INSERM U1015, Université Paris-Saclay, Villejuif, France
| | - Philippe Dessen
- Bioinformatics Platform, UMS AMMICA, CNRS, INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Guillaume Meurice
- Bioinformatics Platform, UMS AMMICA, CNRS, INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Morgane Le-Gall
- Proteom'IC facility, Université Paris Cité, CNRS, INSERM, Institut Cochin, F-75014, Paris, France
| | - Melanie Polrot
- Pre-clinical Evaluation Unit (PFEP), INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Karine Ser-Le Roux
- Pre-clinical Evaluation Unit (PFEP), INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Kamel Mamchaoui
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, F-75013, Paris, France
| | - Nathalie Droin
- Genomic Platform, UMS AMMICA US 23 INSERM UAR 3655 CNRS, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
- UMR1287 INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Hana Raslova
- UMR1287 INSERM, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France
| | - Pascal Maire
- INSERM U1016, CNRS UMR 8104, Institut Cochin, Université Paris-Cité, Paris, France
| | - Birgit Geoerger
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer campus, INSERM U1015, Université Paris-Saclay, Villejuif, France
| | - Iryna Pirozhkova
- UMR8126 CNRS, Gustave Roussy Cancer campus, Université Paris-Saclay, Villejuif, France.
- INSERM U1016, CNRS UMR 8104, Institut Cochin, Université Paris-Cité, Paris, France.
| |
Collapse
|
10
|
Toyoda M, Fukuda T, Fujimoto R, Kawakami K, Hayashi C, Nakao Y, Watanabe Y, Aoki T, Shida M, Sanui T, Taguchi M, Yamamichi K, Okabe A, Okada T, Oka K, Nakayama K, Nishimura F, Kajioka S. Scaffold-free bone-like 3D structure established through osteogenic differentiation from human gingiva-derived stem cells. Biochem Biophys Rep 2024; 38:101656. [PMID: 38379857 PMCID: PMC10878834 DOI: 10.1016/j.bbrep.2024.101656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/02/2024] [Accepted: 01/27/2024] [Indexed: 02/22/2024] Open
Abstract
Introduction & objectives Stem cell therapy for regenerative medicine has been sincerely investigated, but not still popular although some clinical trials show hopeful results. This therapy is suggested to be a representative candidate such as bone defect due to the accident, iatrogenic resection oncological tumor, congenital disease, and severe periodontitis in oral region. Recently, the Bio-3D printer "Regenova®" has been introduced as an innovative three-dimensional culture system, equipped scaffold-free bio-assembling techniques without any biomaterials. Therefore, we expected a mount of bone defect could be repaired by the structure established from this Bio-3D printer using osteogenic potential stem cells. Material & methods The gingival tissue (1x1 mm) was removed from the distal part of the lower wisdom tooth of the patients who agreed our study. Human Gingival Mesenchymal Stem Cells (hGMSCs) were isolated from this tissue and cultured, since we confirmed the characteristics such as facile isolation and accelerated proliferation, further, strong potential of osteogenic-differentiation. Spheroids were formed using hGMSC in 96-well plates designed for low cell adhesion. The size of the spheroids was measured, and fluorescent immunostaining was employed to verify the expression of stem cell and apoptosis marker, and extracellular matrix. Following four weeks of bone differentiation, μCT imaging was performed. Calcification was confirmed by alizarin red and von Kossa staining. Fluorescent immunostaining was utilized to assess the expression of markers indicative of advanced bone differentiation. Results We have established and confirmed the spheroids (∼600 μm in diameter) constructed from human GMSCs (hGMSCs) still maintain stem cell potentials and osteogenic differentiation abilities from the results that CD73 and not CD34 were expressed as stem cell positive and negative marker, respectively. These spheroids were pilled up like cylindal shape to the "Kenzan" platform of Bio-3D printer and cultured for 7days. The cylindal structure originated from compound spheroids were tried to differentiate into bone four weeks with osteogenic induction medium. The calcification of bio-3D printed bone-like structures was confirmed by alizarin red and Von Kossa staining. In addition, μCT analysis revealed that the HU (Hounsfield Unit) of the calcified structures was almost identical to that of trabecular bone. Immunofluorescent staining detected osteocalcin expression, a late-stage bone differentiation marker. Conclusion For the first time, we have achieved the construction of a scaffold-free, bone-like luminal structure through the assembly of spheroids comprised of this hGMSCs. This success is sure to be close to the induction of clinical application against regenerative medicine especially for bone defect disease.
Collapse
Affiliation(s)
- Masaaki Toyoda
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Takao Fukuda
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Ryota Fujimoto
- Center for Regenerative Medicine Research, Faculty of Medicine, Saga University, Saga, Japan
| | - Kentaro Kawakami
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Chikako Hayashi
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yuki Nakao
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yukari Watanabe
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Tsukasa Aoki
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Miyu Shida
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Terukazu Sanui
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Masahide Taguchi
- Section of Pediatric Dentistry, Department of Oral Growth and Development, Fukuoka Dental College, Fukuoka, Japan
| | - Kensuke Yamamichi
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Ayami Okabe
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tatsunori Okada
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kyoko Oka
- Section of Pediatric Dentistry, Department of Oral Growth and Development, Fukuoka Dental College, Fukuoka, Japan
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
| | - Koichi Nakayama
- Center for Regenerative Medicine Research, Faculty of Medicine, Saga University, Saga, Japan
| | - Fusanori Nishimura
- Department of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Shunichi Kajioka
- Department of Pharmaceutical Sciences, International University of Health and Welfare, Fukuoka, Japan
| |
Collapse
|
11
|
Malinauskas T, Moore G, Rudolf AF, Eggington H, Belnoue-Davis HL, El Omari K, Griffiths SC, Woolley RE, Duman R, Wagner A, Leedham SJ, Baldock C, Ashe HL, Siebold C. Molecular mechanism of BMP signal control by Twisted gastrulation. Nat Commun 2024; 15:4976. [PMID: 38862520 PMCID: PMC11167000 DOI: 10.1038/s41467-024-49065-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 05/22/2024] [Indexed: 06/13/2024] Open
Abstract
Twisted gastrulation (TWSG1) is an evolutionarily conserved secreted glycoprotein which controls signaling by Bone Morphogenetic Proteins (BMPs). TWSG1 binds BMPs and their antagonist Chordin to control BMP signaling during embryonic development, kidney regeneration and cancer. We report crystal structures of TWSG1 alone and in complex with a BMP ligand, Growth Differentiation Factor 5. TWSG1 is composed of two distinct, disulfide-rich domains. The TWSG1 N-terminal domain occupies the BMP type 1 receptor binding site on BMPs, whereas the C-terminal domain binds to a Chordin family member. We show that TWSG1 inhibits BMP function in cellular signaling assays and mouse colon organoids. This inhibitory function is abolished in a TWSG1 mutant that cannot bind BMPs. The same mutation in the Drosophila TWSG1 ortholog Tsg fails to mediate BMP gradient formation required for dorsal-ventral axis patterning of the early embryo. Our studies reveal the evolutionarily conserved mechanism of BMP signaling inhibition by TWSG1.
Collapse
Affiliation(s)
- Tomas Malinauskas
- Division of Structural Biology, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK.
| | - Gareth Moore
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Amalie F Rudolf
- Division of Structural Biology, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| | - Holly Eggington
- Intestinal Stem Cell Biology Lab, Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
- Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford National Institute for Health Research Biomedical Research Centre, Oxford, UK
| | - Hayley L Belnoue-Davis
- Intestinal Stem Cell Biology Lab, Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
- Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford National Institute for Health Research Biomedical Research Centre, Oxford, UK
| | - Kamel El Omari
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, OX11 0DE, UK
| | - Samuel C Griffiths
- Division of Structural Biology, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Evotec (UK) Ltd., 90 Innovation Drive, Milton Park, Abingdon, OX14 4RZ, UK
| | - Rachel E Woolley
- Division of Structural Biology, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Etcembly Ltd., Atlas Building, Harwell Campus, OX11 0QX, UK
| | - Ramona Duman
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, OX11 0DE, UK
| | - Armin Wagner
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, OX11 0DE, UK
| | - Simon J Leedham
- Intestinal Stem Cell Biology Lab, Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
- Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford National Institute for Health Research Biomedical Research Centre, Oxford, UK
| | - Clair Baldock
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PT, UK
| | - Hilary L Ashe
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK.
| | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK.
| |
Collapse
|
12
|
Shaffer KJ, Smith RAA, Daines AM, Luo X, Lu X, Tan TC, Le BQ, Schwörer R, Hinkley SFR, Tyler PC, Nurcombe V, Cool SM. Rational synthesis of a heparan sulfate saccharide that promotes the activity of BMP2. Carbohydr Polym 2024; 333:121979. [PMID: 38494232 DOI: 10.1016/j.carbpol.2024.121979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/19/2024]
Abstract
Heparan sulfate (HS) is a glycosaminoglycan (GAG) found throughout nature and is involved in a wide range of functions including modulation of cell signalling via sequestration of growth factors. Current consensus is that the specificity of HS motifs for protein binding are individual for each protein. Given the structural complexity of HS the synthesis of libraries of these compounds to probe this is not trivial. Herein we present the synthesis of an HS decamer, the design of which was undertaken rationally from previously published data for HS binding to the growth factor BMP-2. The biological activity of this HS decamer was assessed in vitro, showing that it had the ability to both bind BMP-2 and increase its thermal stability as well as enhancing the bioactivity of BMP-2 in vitro in C2C12 cells. At the same time no undesired anticoagulant effect was observed. This decamer was then analysed in vivo in a rabbit model where higher bone formation, bone mineral density (BMD) and trabecular thickness were observed over an empty defect or collagen implant alone. This indicated that the HS decamer was effective in promoting bone regeneration in vivo.
Collapse
Affiliation(s)
- Karl J Shaffer
- Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Road, Gracefield, Lower Hutt 5010, New Zealand
| | - Raymond A A Smith
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138673, Singapore; School of Chemical Engineering, University of Queensland, Brisbane, Qld 4072, Australia
| | - Alison M Daines
- Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Road, Gracefield, Lower Hutt 5010, New Zealand.
| | - Xiaoman Luo
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| | - Xiaohua Lu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| | - Tuan Chun Tan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| | - Bach Q Le
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138673, Singapore
| | - Ralf Schwörer
- Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Road, Gracefield, Lower Hutt 5010, New Zealand
| | - Simon F R Hinkley
- Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Road, Gracefield, Lower Hutt 5010, New Zealand
| | - Peter C Tyler
- Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Road, Gracefield, Lower Hutt 5010, New Zealand
| | - Victor Nurcombe
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 138632, Singapore
| | - Simon M Cool
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138673, Singapore; School of Chemical Engineering, University of Queensland, Brisbane, Qld 4072, Australia; Department of Orthopaedic Surgery, Yong Yoo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
13
|
Ding R, Xi Y, Ito A, Shimizu K, Nagamori E, Fujita H, Kawamoto T, Horie M. Bone morphogenetic protein signaling inhibitor improves differentiation and function of 3D muscle construct fabricated using C2C12. J Biosci Bioeng 2024; 137:480-486. [PMID: 38604883 DOI: 10.1016/j.jbiosc.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/14/2024] [Accepted: 03/23/2024] [Indexed: 04/13/2024]
Abstract
Functional tissue-engineered artificial skeletal muscle tissue has great potential for pharmacological and academic applications. This study demonstrates an in vitro tissue engineering system to construct functional artificial skeletal muscle tissues using self-organization and signal inhibitors. To induce efficient self-organization, we optimized the substrate stiffness and extracellular matrix (ECM) coatings. We modified the tissue morphology to be ring-shaped under optimized self-organization conditions. A bone morphogenetic protein (BMP) inhibitor was added to improve overall myogenic differentiation. This supplementation enhanced the myogenic differentiation ratio and myotube hypertrophy in two-dimensional cell cultures. Finally, we found that myotube hypertrophy was enhanced by a combination of self-organization with ring-shaped tissue and a BMP inhibitor. BMP inhibitor treatment significantly improved myogenic marker expression and contractile force generation in the self-organized tissue. These observations indicated that this procedure may provide a novel and functional artificial skeletal muscle for pharmacological studies.
Collapse
Affiliation(s)
- Ran Ding
- Graduate School of Human and Environmental, Kyoto University, Yoshida-Konoe-Cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yuan Xi
- Graduate School of Human and Environmental, Kyoto University, Yoshida-Konoe-Cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Akira Ito
- Department of Chemical Systems Engineering, School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Kazunori Shimizu
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya 464-8603, Japan
| | - Eiji Nagamori
- Department of Biomedical Engineering, Osaka Institute of Technology, 5-16-1 Omiya, Asahi-ku, Osaka 535-8585, Japan
| | - Hideaki Fujita
- Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Takuo Kawamoto
- Graduate School of Human and Environmental, Kyoto University, Yoshida-Konoe-Cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Masanobu Horie
- Division of Biochemical Engineering, Radioisotope Research Center, Kyoto University, Yoshida-Konoe-Cho, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
14
|
Liu Y, Li Y, Liu Y, Gao Z, Zhang J, Qiu Y, Wang C, Lu X, Yang J. Investigation of the Shared Biomarkers in Heterotopic Ossification Between Ossification of the Ligamentum Flavum and Ankylosing Spondylitis. Global Spine J 2024:21925682241255894. [PMID: 38757696 DOI: 10.1177/21925682241255894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/18/2024] Open
Abstract
STUDY DESIGN Bioinformatics analysis of Gene Expression Omnibus (GEO). OBJECTIVE Ossification of the ligamentum flavum (OLF) and ankylosing spondylitis (AS) represent intricate conditions marked by the gradual progression of endochondral ossification. This investigation endeavors to unveil common biomarkers associated with heterotopic ossification and explore the potential molecular regulatory mechanisms. METHODS Microarray and RNA-sequencing datasets retrieved from the Gene Expression Omnibus (GEO) repository were harnessed to discern differentially expressed genes (DEGs) within the OLF and AS datasets. Subsequently, Weighted Gene Co-expression Network Analysis (WGCNA) was implemented to pinpoint co-expression modules linked to OLF and AS. Common genes were further subjected to an examination of functional pathway enrichment. Moreover, hub intersection genes were identified using the Least Absolute Shrinkage and Selection Operator (LASSO) regression, followed by an evaluation of diagnostic performance in external OLF and AS cohorts. Lastly, an analysis of immune cell infiltration was conducted to scrutinize the correlation of immune cell presence with shared biomarkers in OLF and AS. RESULTS A total of 1353 and 91 Differentially Expressed Genes (DEGs) were identified in OLF and AS, respectively. Using the Weighted Gene Co-expression Network Analysis (WGCNA), 2 modules were found to be notably significant for OLF and AS. The integrative bioinformatic analysis revealed 3 hub genes (MAB21L2, MEGF10, ISLR) as shared risk biomarkers, with MAB21L2 being the central focus. Receiver Operating Characteristic (ROC) analysis exhibited a strong diagnostic potential for these hub genes. Gene Ontology (GO) analysis indicated their involvement in the positive regulation of myoblast proliferation. Notably, MAB21L2 was singled out as the optimal common biomarker for OLF and AS. Furthermore, an analysis of immune infiltration demonstrated a correlation between MAB21L2 expression and changes in immune cells. Activated CD8 T cells were identified as shared differential immune infiltrating cells significantly linked to MAB21L2 in both OLF and AS. CONCLUSION This study represents the first instance of identifying MAB21L2 as a prospective diagnostic marker for patients contending with OLF associated with AS. The research results indicate that the ECM-receptor interaction and the cell-cell adhesion may play a role in both disease processes. This newfound knowledge not only enhances our understanding of the pathogenesis behind spinal ligament ossification but also uncovers potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Yishan Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, People's Republic of China
- Department of Orthopaedic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
- Department of Spinal Surgery, Subei People's Hospital, Clinical Medical School, Yangzhou University Affiliated Hospital, Yangzhou, China
| | - Yang Li
- Department of Orthopaedic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yixuan Liu
- Department of Spinal Surgery, Subei People's Hospital, Clinical Medical School, Yangzhou University Affiliated Hospital, Yangzhou, China
- Dalian Medical University, Dalian, China
| | - Zhongya Gao
- Department of Orthopaedic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jianjun Zhang
- Department of Orthopaedic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
- North Sichuan Medical College, Nanchong, China
| | - Youcai Qiu
- Department of Orthopaedic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Can Wang
- Department of Orthopaedic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
- North Sichuan Medical College, Nanchong, China
| | - Xuhua Lu
- Department of Orthopaedic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jiandong Yang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, People's Republic of China
- Department of Spinal Surgery, Subei People's Hospital, Clinical Medical School, Yangzhou University Affiliated Hospital, Yangzhou, China
| |
Collapse
|
15
|
Ito T, Rojasawasthien T, Takeuchi SY, Okamoto H, Okumura N, Shirakawa T, Matsubara T, Kawamoto T, Kokabu S. Royal Jelly Enhances the Ability of Myoblast C2C12 Cells to Differentiate into Multilineage Cells. Molecules 2024; 29:1449. [PMID: 38611729 PMCID: PMC11013243 DOI: 10.3390/molecules29071449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024] Open
Abstract
Royal jelly (RJ) is recognized as beneficial to mammalian health. Multilineage differentiation potential is an important property of mesenchymal stem cells (MSCs). C2C12 cells have an innate ability to differentiate into myogenic cells. Like MSCs, C2C12 cells can also differentiate into osteoblast- and adipocyte-lineage cells. We recently reported that RJ enhances the myogenic differentiation of C2C12 cells. However, the effect of RJ on osteoblast or adipocyte differentiation is still unknown. Here in this study, we have examined the effect of RJ on the osteoblast and adipocyte differentiation of C2C12 cells. Protease-treated RJ was used to reduce the adverse effects caused by RJ supplementation. To induce osteoblast or adipocyte differentiation, cells were treated with bone morphogenetic proteins (BMP) or peroxisome proliferator-activated receptor γ (PPARγ) agonist, respectively. RNA-seq was used to analyze the effect of RJ on gene expression. We found that RJ stimulates osteoblast and adipocyte differentiation. RJ regulated 279 genes. RJ treatment upregulated glutathione-related genes. Glutathione, the most abundant antioxidative factor in cells, has been shown to promote osteoblast differentiation in MSC and MSC-like cells. Therefore, RJ may promote osteogenesis, at least in part, through the antioxidant effects of glutathione. RJ enhances the differentiation ability of C2C12 cells into multiple lineages, including myoblasts, osteoblasts, and adipocytes.
Collapse
Affiliation(s)
- Takumi Ito
- Division of Molecular Signaling and Biochemistry, Kyushu Dental University, Fukuoka 803-8580, Japan; (T.I.); (T.R.); (S.Y.T.); (T.M.)
- Division of Orofacial Functions and Orthodontics, Kyushu Dental University, Fukuoka 803-8580, Japan; (T.S.); (T.K.)
| | - Thira Rojasawasthien
- Division of Molecular Signaling and Biochemistry, Kyushu Dental University, Fukuoka 803-8580, Japan; (T.I.); (T.R.); (S.Y.T.); (T.M.)
| | - Sachiko Yamashita Takeuchi
- Division of Molecular Signaling and Biochemistry, Kyushu Dental University, Fukuoka 803-8580, Japan; (T.I.); (T.R.); (S.Y.T.); (T.M.)
| | - Hideto Okamoto
- Institute for Bee Products and Health Science, Yamada Bee Company, Inc., Okayama 708-0393, Japan; (H.O.); (N.O.)
| | - Nobuaki Okumura
- Institute for Bee Products and Health Science, Yamada Bee Company, Inc., Okayama 708-0393, Japan; (H.O.); (N.O.)
| | - Tomohiko Shirakawa
- Division of Orofacial Functions and Orthodontics, Kyushu Dental University, Fukuoka 803-8580, Japan; (T.S.); (T.K.)
| | - Takuma Matsubara
- Division of Molecular Signaling and Biochemistry, Kyushu Dental University, Fukuoka 803-8580, Japan; (T.I.); (T.R.); (S.Y.T.); (T.M.)
| | - Tatsuo Kawamoto
- Division of Orofacial Functions and Orthodontics, Kyushu Dental University, Fukuoka 803-8580, Japan; (T.S.); (T.K.)
| | - Shoichiro Kokabu
- Division of Molecular Signaling and Biochemistry, Kyushu Dental University, Fukuoka 803-8580, Japan; (T.I.); (T.R.); (S.Y.T.); (T.M.)
| |
Collapse
|
16
|
Juan C, Bancroft AC, Choi JH, Nunez JH, Pagani CA, Lin YS, Hsiao EC, Levi B. Intersections of Fibrodysplasia Ossificans Progressiva and Traumatic Heterotopic Ossification. Biomolecules 2024; 14:349. [PMID: 38540768 PMCID: PMC10968060 DOI: 10.3390/biom14030349] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/29/2024] [Accepted: 03/08/2024] [Indexed: 11/11/2024] Open
Abstract
Heterotopic ossification (HO) is a debilitating pathology where ectopic bone develops in areas of soft tissue. HO can develop as a consequence of traumatic insult or as a result of dysregulated osteogenic signaling, as in the case of the orphan disease fibrodysplasia ossificans progressiva (FOP). Traumatic HO (tHO) formation is mediated by the complex interplay of signaling between progenitor, inflammatory, and nerve cells, among others, making it a challenging process to understand. Research into the pathogenesis of genetically mediated HO (gHO) in FOP has established a pathway involving uninhibited activin-like kinase 2 receptor (ALK2) signaling that leads to downstream osteogenesis. Current methods of diagnosis and treatment lag behind pre-mature HO detection and progressive HO accumulation, resulting in irreversible decreases in range of motion and chronic pain for patients. As such, it is necessary to draw on advancements made in the study of tHO and gHO to better diagnose, comprehend, prevent, and treat both.
Collapse
Affiliation(s)
- Conan Juan
- Center for Organogenesis, Regeneration, and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA (J.H.C.)
| | - Alec C. Bancroft
- Center for Organogenesis, Regeneration, and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA (J.H.C.)
- Baylor College of Medicine, Houston, TX 77030, USA
| | - Ji Hae Choi
- Center for Organogenesis, Regeneration, and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA (J.H.C.)
| | - Johanna H. Nunez
- Center for Organogenesis, Regeneration, and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA (J.H.C.)
| | - Chase A. Pagani
- Center for Organogenesis, Regeneration, and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA (J.H.C.)
| | - Yen-Sheng Lin
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Edward C. Hsiao
- Division of Endocrinology and Metabolism, Department of Medicine, the Institute for Human Genetics, and the Program in Craniofacial Biology, University of California San Francisco Medical Center, San Francisco, CA 94143, USA;
| | - Benjamin Levi
- Center for Organogenesis, Regeneration, and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA (J.H.C.)
| |
Collapse
|
17
|
Vallmajo-Martin Q, Millan C, Müller R, Weber FE, Ehrbar M, Ghayor C. Enhanced bone regeneration in rat calvarial defects through BMP2 release from engineered poly(ethylene glycol) hydrogels. Sci Rep 2024; 14:4916. [PMID: 38418564 PMCID: PMC10901800 DOI: 10.1038/s41598-024-55411-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 02/23/2024] [Indexed: 03/01/2024] Open
Abstract
The clinical standard therapy for large bone defects, typically addressed through autograft or allograft donor tissue, faces significant limitations. Tissue engineering offers a promising alternative strategy for the regeneration of substantial bone lesions. In this study, we harnessed poly(ethylene glycol) (PEG)-based hydrogels, optimizing critical parameters including stiffness, incorporation of arginine-glycine-aspartic acid (RGD) cell adhesion motifs, degradability, and the release of BMP2 to promote bone formation. In vitro we demonstrated that human bone marrow derived stromal cell (hBMSC) proliferation and spreading strongly correlates with hydrogel stiffness and adhesion to RGD peptide motifs. Moreover, the incorporation of the osteogenic growth factor BMP2 into the hydrogels enabled sustained release, effectively inducing bone regeneration in encapsulated progenitor cells. When used in vivo to treat calvarial defects in rats, we showed that hydrogels of low and intermediate stiffness optimally facilitated cell migration, proliferation, and differentiation promoting the efficient repair of bone defects. Our comprehensive in vitro and in vivo findings collectively suggest that the developed hydrogels hold significant promise for clinical translation for bone repair and regeneration by delivering sustained and controlled stimuli from active signaling molecules.
Collapse
Affiliation(s)
- Queralt Vallmajo-Martin
- Department of Obstetrics, University Hospital Zürich, University of Zürich, Schmelzbergstrasse 12, 8091, Zurich, Switzerland
- School of Life Sciences and School of Engineering, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Station 15, 1015, Lausanne, Switzerland
| | - Christopher Millan
- Department of Urology, University Hospital Zürich, University of Zürich, Wagistrasse 21, 8952, Zurich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, Eidgenössische Technische Hochschule Zürich, Leopold-Ruzicka-Weg 8093, 8049, Zurich, Switzerland
| | - Franz E Weber
- Center of Dental Medicine, Oral Biotechnology & Bioengineering, University of Zürich, Plattenstrasse 11, 8032, Zurich, Switzerland
| | - Martin Ehrbar
- Department of Obstetrics, University Hospital Zürich, University of Zürich, Schmelzbergstrasse 12, 8091, Zurich, Switzerland.
| | - Chafik Ghayor
- Center of Dental Medicine, Oral Biotechnology & Bioengineering, University of Zürich, Plattenstrasse 11, 8032, Zurich, Switzerland.
| |
Collapse
|
18
|
Wagner J, Luck S, Loger K, Açil Y, Spille JH, Kurz S, Ahlhelm M, Schwarzer-Fischer E, Ingwersen LC, Jonitz-Heincke A, Sedaghat S, Wiltfang J, Naujokat H. Bone regeneration in critical-size defects of the mandible using biomechanically adapted CAD/CAM hybrid scaffolds: An in vivo study in miniature pigs. J Craniomaxillofac Surg 2024; 52:127-135. [PMID: 38129185 DOI: 10.1016/j.jcms.2023.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
The study aimed to analyze bone regeneration in critical-size defects using hybrid scaffolds biomechanically adapted to the specific defect and adding the growth factor rhBMP-2. For this animal study, ten minipigs underwent bilateral defects in the corpus mandibulae and were subsequently treated with novel cylindrical hybrid scaffolds. These scaffolds were designed digitally to suit the biomechanical requirements of the mandibular defect, utilizing finite element analysis. The scaffolds comprised zirconium dioxide-tricalcium phosphate (ZrO2-TCP) support struts and TCP foam ceramics. One scaffold in each animal was loaded with rhBMP-2 (100 μg/cm³), while the other served as an unloaded negative control. Fluorescent dyes were administered every 2 weeks, and computed tomography (CT) scans were conducted every 4 weeks. Euthanasia was performed after 3 months, and samples were collected for examination using micro-CT and histological evaluation of both hard and soft tissue. Intravital CT examinations revealed minor changes in radiographic density from 4 to 12 weeks postoperatively. In the group treated with rhBMP-2, radiographic density shifted from 2513 ± 128 (mean ± SD) to 2606 ± 115 Hounsfield units (HU), while the group without rhBMP-2 showed a change from 2430 ± 131 to 2601 ± 67 HU. Prior to implantation, the radiological density of samples measured 1508 ± 30 mg HA/cm³, whereas post-mortem densities were 1346 ± 71 mg HA/cm³ in the rhBMP-2 group and 1282 ± 91 mg HA/cm³ in the control group (p = 0.045), as indicated by micro-CT measurements. The histological assessment demonstrated successful ossification in all specimens. The newly formed bone area proportion was significantly greater in the rhBMP-2 group (48 ± 10%) compared with the control group without rhBMP-2 (42 ± 9%, p = 0.03). The mean area proportion of remaining TCP foam was 23 ± 8% with rhBMP-2 and 24 ± 10% without rhBMP-2. Successful bone regeneration was accomplished by implanting hybrid scaffolds into critical-size mandibular defects. Loading these scaffolds with rhBMP-2 led to enhanced bone regeneration and a uniform distribution of new bone formation within the hybrid scaffolds. Further studies are required to determine the adaptability of hybrid scaffolds for larger and potentially segmental defects in the maxillofacial region.
Collapse
Affiliation(s)
- Juliane Wagner
- Department of Oral and Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany; Cluster of Excellence, Precision Medicine in Inflammation, Christian-Albrechts-University of Kiel, Kiel, Germany.
| | - Sascha Luck
- Department of Oral and Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Klaas Loger
- Department of Oral and Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Yahya Açil
- Department of Oral and Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Johannes H Spille
- Department of Oral and Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Sascha Kurz
- ZESBO - Center for Research on Musculoskeletal Systems, Leipzig University, Leipzig, Germany
| | - Matthias Ahlhelm
- Fraunhofer Institute for Ceramic Technologies and Systems, IKTS, Dresden, Germany
| | | | - Lena-Christin Ingwersen
- Biomechanics and Implant Technology Research Laboratory, Department of Orthopaedics, Rostock University Medical Center, Rostock, Germany
| | - Anika Jonitz-Heincke
- Biomechanics and Implant Technology Research Laboratory, Department of Orthopaedics, Rostock University Medical Center, Rostock, Germany
| | - Sam Sedaghat
- Department of Diagnostic and Interventional Radiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Jörg Wiltfang
- Department of Oral and Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Hendrik Naujokat
- Department of Oral and Maxillofacial Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
19
|
Zhou L, Wang J, Mu W. BMP-2 promotes fracture healing by facilitating osteoblast differentiation and bone defect osteogenesis. Am J Transl Res 2023; 15:6751-6759. [PMID: 38187002 PMCID: PMC10767540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 11/28/2023] [Indexed: 01/09/2024]
Abstract
OBJECTIVE To investigate the role of bone morphogenetic protein-2 (BMP-2) in promoting fracture healing in animal models. METHODS Mouse models with muscle bag heterotopic osteogenesis (HO) were divided into a HO control group (not implanted with 250 μg rhBMP-2 bone repairing material), and a HO observation group (implanted with 250 μg rhBMP-2 bone repairing material); while rat models with bone defect (BD) were divided into a BD control group (not implanted with 250 μg rhBMP-2 bone repairing material) and a BD observation group (implanted with 250 μg rhBMP-2 bone repairing material). At 4 weeks after HO establishment, the new bone formation at the operation site was observed through visual inspections and X-ray scanning. The content of serum alkaline phosphatase (ALP) was detected by automatic biochemical analyzer. The formation of new bone at the operative sites was observed by Hematoxylin and eosin staining and Masson staining. At 0, 2, 4 and 8 weeks after operation, the growth of the defect area and its surrounding callus were observed by X-ray scanning. At 4 and 8 weeks after bone defect establishment in the mouse models, the histological changes and osteogenesis of the bone defect site were observed. RESULTS The heterotopic osteogenesis experiment showed that at 4 weeks after operation, the mass at the muscle bag in the HO observation group became larger in contrast to the HO control group. X-ray scanning showed that there was obvious irregular bone shadow at the back muscle bag of mice from the HO observation group. The content of serum ALP in the HO observation group was significantly higher than that in the HO control group (all P<0.05). The muscle pocket in the HO observation group showed higher ectopic osteogenic activity comparing with the HO control group. Histological staining showed that bone tissue structure was visible in the newly regenerated bone, forming bone trabeculae and bone marrow tissue. Under the microscope, a large number of osteoblasts arranged neatly in a cubic shape presented at the edge of the new bone, and there were bone lacunae formed, and the bone tissue was in a relatively mature stage. In the rat bone defect models, X-ray scanning showed that the high-density development area was further increased. There was a large amount of callus formation in the bone defect area of the BD observation group, while the BD control group still had no high-density development. At 8 weeks after operation, the high-density development area decreased, indicating that there was partial absorption of callus, while there was still no high-density development in the BD control group. The callus of the bone defect area in the BD observation group was reduced and the defect area was gradually repaired, while the bone defect in the BD control group was still obvious and the bone repair was not completed. CONCLUSIONS BMP-2 could promote osteoblast differentiation and bone defect osteogenesis in vivo. Thus, it is worthy of clinical application.
Collapse
Affiliation(s)
- Lei Zhou
- Department of Orthopaedics, Shandong Provincial Hospital, Shandong UniversityJinan, Shandong, China
- Department of Orthopaedics, The Second Affiliated Hospital of Xuzhou Medical UniversityXuzhou, Jiangsu, China
| | - Jianqiang Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Xuzhou Medical UniversityXuzhou, Jiangsu, China
| | - Weidong Mu
- Department of Orthopaedics, Shandong Provincial Hospital, Shandong UniversityJinan, Shandong, China
| |
Collapse
|
20
|
Ding S, Ma Y, Yang J, Tang Y, Jin Y, Li L, Ma C. MiR-224-5p inhibits osteoblast differentiation and impairs bone formation by targeting Runx2 and Sp7. Cytotechnology 2023; 75:505-516. [PMID: 37841957 PMCID: PMC10575840 DOI: 10.1007/s10616-023-00593-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 08/24/2023] [Indexed: 10/17/2023] Open
Abstract
Osteoporosis is a complicated multifactorial disorder characterized by low bone mass and deteriorated bone microarchitecture with an elevated fracture risk. MicroRNAs play important roles in osteoblastic differentiation. In the present study, we found that miR-224-5p was markedly downregulated during the osteogenic differentiation of C2C12 cells. Overexpression of miR-224-5p in C2C12 cells inhibited osteoblast activity, as indicated by reduced ALP activity, matrix mineralization and the expression of osteogenic marker genes. Moreover, we demonstrated that Runx2 and Sp7 were direct targets of miR-224-5p. Furthermore, the specific inhibition of miR-224-5p by femoral bone marrow cavity injection with miR-224-5p antagomir prevented ovariectomy-induced bone loss. Finally, we found that the levels of miR-224-5p were markedly elevated in the sera of patients with osteoporosis. Collectively, this study revealed that miR-224-5p negatively regulates osteogenic differentiation by targeting Runx2 and Sp7. It also highlights the potential use of miR-224-5p as a therapeutic target and diagnostic biomarker for osteoporosis. Supplementary information The online version contains supplementary material available at 10.1007/s10616-023-00593-z.
Collapse
Affiliation(s)
- Siyang Ding
- Department of Medical Genetics, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166 China
- Jiangsu Key Laboratory of Oral Disease, Department of Sixth Outpatient, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, 210029 China
| | - Yunfei Ma
- Department of Medical Genetics, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166 China
| | - Jiashu Yang
- Department of Medical Genetics, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166 China
| | - Yuting Tang
- Department of Medical Genetics, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166 China
| | - Yucui Jin
- Department of Medical Genetics, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166 China
| | - Lingyun Li
- Department of Medical Genetics, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166 China
| | - Changyan Ma
- Department of Medical Genetics, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166 China
| |
Collapse
|
21
|
Yuan M, Liu X, Wang M, Li Z, Li H, Leng L, Wang S. A Functional Variant Alters the Binding of Bone morphogenetic protein 2 to the Transcription Factor NF-κB to Regulate Bone morphogenetic protein 2 Gene Expression and Chicken Abdominal Fat Deposition. Animals (Basel) 2023; 13:3401. [PMID: 37958155 PMCID: PMC10650395 DOI: 10.3390/ani13213401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
In this study, we employed a dual-luciferase reporter assay and electrophoretic mobility shift analysis (EMSA) in vitro to explore whether a 12-base pair (bp) insertion/deletion (InDel) variant (namely g.14798187_14798188insTCCCTGCCCCCT) within intron 2 of the chicken BMP2 gene, which was significantly associated with chicken abdominal fat weight and abdominal fat percentage, is a functional marker and its potential regulatory mechanism. The reporter analysis demonstrated that the luciferase activity of the deletion allele was extremely significantly higher than that of the insertion allele (p < 0.01). A bioinformatics analysis revealed that compared to the deletion allele, the insertion allele created a transcription factor binding site of nuclear factor-kappa B (NF-κB), which exhibited an inhibitory effect on fat deposition. A dual-luciferase reporter assay demonstrated that the inhibitory effect of NF-κB on the deletion allele was stronger than that on the insertion allele. EMSA indicated that the binding affinity of NF-κB for the insertion allele was stronger than that for the deletion allele. In conclusion, the 12-bp InDel chicken BMP2 gene variant is a functional variant affecting fat deposition in chickens, which may partially regulate BMP2 gene expression by affecting the binding of transcription factor NF-κB to the BMP2 gene.
Collapse
Affiliation(s)
- Meng Yuan
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China; (M.Y.); (X.L.); (M.W.); (Z.L.); (H.L.)
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Xin Liu
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China; (M.Y.); (X.L.); (M.W.); (Z.L.); (H.L.)
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Mengdie Wang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China; (M.Y.); (X.L.); (M.W.); (Z.L.); (H.L.)
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Ziwei Li
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China; (M.Y.); (X.L.); (M.W.); (Z.L.); (H.L.)
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Hui Li
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China; (M.Y.); (X.L.); (M.W.); (Z.L.); (H.L.)
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Li Leng
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China; (M.Y.); (X.L.); (M.W.); (Z.L.); (H.L.)
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Shouzhi Wang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China; (M.Y.); (X.L.); (M.W.); (Z.L.); (H.L.)
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
22
|
Siverino C, Fahmy-Garcia S, Niklaus V, Kops N, Dolcini L, Misciagna MM, Ridwan Y, Farrell E, van Osch GJ, Nickel J. Addition of heparin binding sites strongly increases the bone forming capabilities of BMP9 in vivo. Bioact Mater 2023; 29:241-250. [PMID: 37502679 PMCID: PMC10371762 DOI: 10.1016/j.bioactmat.2023.07.010] [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: 03/09/2023] [Revised: 06/29/2023] [Accepted: 07/09/2023] [Indexed: 07/29/2023] Open
Abstract
Bone Morphogenetic proteins (BMPs) like BMP2 and BMP7 have shown great potential in the treatment of severe bone defects. In recent in vitro studies, BMP9 revealed the highest osteogenic potential compared to other BMPs, possibly due to its unique signaling pathways that differs from other osteogenic BMPs. However, in vivo the bone forming capacity of BMP9-adsorbed scaffolds is not superior to BMP2 or BMP7. In silico analysis of the BMP9 protein sequence revealed that BMP9, in contrast to other osteogenic BMPs such as BMP2, completely lacks so-called heparin binding motifs that enable extracellular matrix (ECM) interactions which in general might be essential for the BMPs' osteogenic function. Therefore, we genetically engineered a new BMP9 variant by adding BMP2-derived heparin binding motifs to the N-terminal segment of BMP9's mature part. The resulting protein (BMP9 HB) showed higher heparin binding affinity than BMP2, similar osteogenic activity in vitro and comparable binding affinities to BMPR-II and ALK1 compared to BMP9. However, remarkable differences were observed when BMP9 HB was adsorbed to collagen scaffolds and implanted subcutaneously in the dorsum of rats, showing a consistent and significant increase in bone volume and density compared to BMP2 and BMP9. Even at 10-fold lower BMP9 HB doses bone tissue formation was observed. This innovative approach of significantly enhancing the osteogenic properties of BMP9 simply by addition of ECM binding motifs, could constitute a valuable replacement to the commonly used BMPs. The possibility to use lower protein doses demonstrates BMP9 HB's high translational potential.
Collapse
Affiliation(s)
- Claudia Siverino
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Shorouk Fahmy-Garcia
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Viktoria Niklaus
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Nicole Kops
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Laura Dolcini
- Fin-Ceramica Faenza SpA, Via Granarolo 177/3, 48018, Faenza, Italy
| | | | - Yanto Ridwan
- AMIE Core Facility, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Gerjo J.V.M. van Osch
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime and Materials Engineering, Delft University of Technology, Delft, the Netherlands
| | - Joachim Nickel
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
- Fraunhofer ISC, Translational Center RT, Wuerzburg, Germany
| |
Collapse
|
23
|
Piao M, Lee SH, Li Y, Choi JK, Yeo CY, Lee KY. Cyclophilin E (CypE) Functions as a Positive Regulator in Osteoblast Differentiation by Regulating the Transcriptional Activity of Runx2. Cells 2023; 12:2549. [PMID: 37947627 PMCID: PMC10648996 DOI: 10.3390/cells12212549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 10/20/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023] Open
Abstract
Cyclophilin E (CypE) belongs to the cyclophilin family and exhibits peptidyl-prolyl cis-trans isomerase (PPIase) activity. It participates in various biological processes through the regulation of peptidyl-prolyl isomerization. However, the specific role of CypE in osteoblast differentiation has not yet been elucidated. In this study, we first discovered the positive impact of CypE on osteoblast differentiation through gain or loss of function experiments. Mechanistically, CypE enhances the transcriptional activity of Runx2 through its PPIase activity. Furthermore, we identified the involvement of the Akt signaling pathway in CypE's function in osteoblast differentiation. Taken together, our findings indicate that CypE plays an important role in osteoblast differentiation as a positive regulator by increasing the transcriptional activity of Runx2.
Collapse
Affiliation(s)
- Meiyu Piao
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; (M.P.); (S.H.L.); (Y.L.)
| | - Sung Ho Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; (M.P.); (S.H.L.); (Y.L.)
| | - Yuankuan Li
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; (M.P.); (S.H.L.); (Y.L.)
| | - Joong-Kook Choi
- Division of Biochemistry, College of Medicine, Chungbuk National University, Cheong-Ju 28644, Republic of Korea;
| | - Chang-Yeol Yeo
- Department of Life Science and Research Center for Cellular Homeostasis, Ewha Woman’s University, Seoul 03760, Republic of Korea
| | - Kwang Youl Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; (M.P.); (S.H.L.); (Y.L.)
| |
Collapse
|
24
|
Lu X, Li L, Wu N, Chen W, Hong S, Xu M, Ding Y, Gao Y. BMP9 functions as a negative regulator in the myogenic differentiation of primary mouse myoblasts. Biosci Biotechnol Biochem 2023; 87:1255-1264. [PMID: 37553201 DOI: 10.1093/bbb/zbad104] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/26/2023] [Indexed: 08/10/2023]
Abstract
BMP9, a member of the TGF-β superfamily, reveals the great translational promise for it has been shown to have the strong effect of osteogenic activity in vitro and in vivo. However, the implantation of certain BMPs (bone morphogenetic proteins) into muscular tissues induces ectopic bone formation. BMPs induce osteoblastic differentiation in skeletal muscle, suggesting that myogenic stem cells, such as myoblasts, are the potential progenitors of osteoblasts during heterotopic bone differentiation. Here, we investigate the role of BMP9 during primary mouse myoblasts differentiation. We found BMP9 enhanced cell proliferation and reduced myogenic differentiation of primary mouse myoblasts. In addition, adenovirus-mediated overexpression of BMP9 delayed muscle regeneration after BaCl2-induced injury. ALK1 knockdown reversed the inhibition of myoblast differentiation induced by BMP9. Our data indicate that BMP9 inhibits myogenic differentiation in primary mouse myoblasts and delays skeletal muscle regeneration after injury.
Collapse
Affiliation(s)
- Xiya Lu
- Department of Dermatology, Shanghai Skin Disease Hospital, Shanghai, China
| | - Liang Li
- Department of Dermatologic Surgery, Shanghai Skin Disease Hospital, Shanghai, China
| | - Nanhui Wu
- Department of Dermatopathology, Shanghai Skin Disease Hospital, Shanghai, China
| | - Wenjuan Chen
- Department of Dermatology, Shanghai Skin Disease Hospital, Shanghai, China
| | - Sheng Hong
- Department of Dermatology, Changhai Hospital, Shanghai, China
| | - Mingyuan Xu
- Department of Dermatopathology, Shanghai Skin Disease Hospital, Shanghai, China
| | - Yangfeng Ding
- Department of Dermatology, Shanghai Skin Disease Hospital, Shanghai, China
| | - Yunlu Gao
- Department of Dermatology, Shanghai Skin Disease Hospital, Shanghai, China
| |
Collapse
|
25
|
Son TH, Kim SH, Shin HL, Kim D, Huh JS, Ryoo R, Choi Y, Choi SW. Inhibition of Osteoclast Differentiation and Promotion of Osteogenic Formation by Wolfiporia extensa Mycelium. J Microbiol Biotechnol 2023; 33:1197-1205. [PMID: 37317624 PMCID: PMC10580891 DOI: 10.4014/jmb.2304.04048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 05/30/2023] [Accepted: 05/30/2023] [Indexed: 06/16/2023]
Abstract
Osteoporosis, Greek for "porous bone," is a bone disease characterized by a decrease in bone strength, microarchitectural changes in the bone tissues, and an increased risk of fracture. An imbalance of bone resorption and bone formation may lead to chronic metabolic diseases such as osteoporosis. Wolfiporia extensa, known as "Bokryung" in Korea, is a fungus belonging to the family Polyporaceae and has been used as a therapeutic food against various diseases. Medicinal mushrooms, mycelium and fungi, possess approximately 130 medicinal functions, including antitumor, immunomodulating, antibacterial, hepatoprotective, and antidiabetic effects, and are therefore used to improve human health. In this study, we used osteoclast and osteoblast cell cultures treated with Wolfiporia extensa mycelium water extract (WEMWE) and investigated the effect of the fungus on bone homeostasis. Subsequently, we assessed its capacity to modulate both osteoblast and osteoclast differentiation by performing osteogenic and anti-osteoclastogenic activity assays. We observed that WEMWE increased BMP-2-stimulated osteogenesis by inducing Smad-Runx2 signal pathway axis. In addition, we found that WEMWE decreased RANKL-induced osteoclastogenesis by blocking c-Fos/NFATc1 via the inhibition of ERK and JNK phosphorylation. Our results show that WEMWE can prevent and treat bone metabolic diseases, including osteoporosis, by a biphasic activity that sustains bone homeostasis. Therefore, we suggest that WEMWE can be used as a preventive and therapeutic drug.
Collapse
Affiliation(s)
- Tae Hyun Son
- School of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
- Forest Biomaterials Research Center, National Institute of Forest Science (NIFoS), Jinju 52817, Republic of Korea
| | - Shin-Hye Kim
- Forest Biomaterials Research Center, National Institute of Forest Science (NIFoS), Jinju 52817, Republic of Korea
| | - Hye-Lim Shin
- Forest Biomaterials Research Center, National Institute of Forest Science (NIFoS), Jinju 52817, Republic of Korea
| | - Dongsoo Kim
- Forest Biomaterials Research Center, National Institute of Forest Science (NIFoS), Jinju 52817, Republic of Korea
| | - Jin-Sung Huh
- Forest Biomaterials Research Center, National Institute of Forest Science (NIFoS), Jinju 52817, Republic of Korea
| | - Rhim Ryoo
- Forest Microbiology Division, Department of Forest Bio-Resources, NIFoS, Suwon 16631, Republic of Korea
| | - Yongseok Choi
- School of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Sik-Won Choi
- Forest Biomaterials Research Center, National Institute of Forest Science (NIFoS), Jinju 52817, Republic of Korea
| |
Collapse
|
26
|
Johnson AL, Kamal M, Parise G. The Role of Supporting Cell Populations in Satellite Cell Mediated Muscle Repair. Cells 2023; 12:1968. [PMID: 37566047 PMCID: PMC10417507 DOI: 10.3390/cells12151968] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/12/2023] Open
Abstract
Skeletal muscle has a high capacity to repair and remodel in response to damage, largely through the action of resident muscle stem cells, termed satellite cells. Satellite cells are required for the proper repair of skeletal muscle through a process known as myogenesis. Recent investigations have observed relationships between satellite cells and other cell types and structures within the muscle microenvironment. These findings suggest that the crosstalk between inflammatory cells, fibrogenic cells, bone-marrow-derived cells, satellite cells, and the vasculature is essential for the restoration of muscle homeostasis. This review will discuss the influence of the cells and structures within the muscle microenvironment on satellite cell function and muscle repair.
Collapse
Affiliation(s)
| | | | - Gianni Parise
- Department of Kinesiology, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
27
|
Padilla-Benavides T, Olea-Flores M, Sharma T, Syed SA, Witwicka H, Zuñiga-Eulogio MD, Zhang K, Navarro-Tito N, Imbalzano AN. Differential Contributions of mSWI/SNF Chromatin Remodeler Sub-Families to Myoblast Differentiation. Int J Mol Sci 2023; 24:11256. [PMID: 37511016 PMCID: PMC10378909 DOI: 10.3390/ijms241411256] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/03/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Mammalian SWI/SNF (mSWI/SNF) complexes are ATP-dependent chromatin remodeling enzymes that are critical for normal cellular functions. mSWI/SNF enzymes are classified into three sub-families based on the presence of specific subunit proteins. The sub-families are Brm- or Brg1-associated factor (BAF), ncBAF (non-canonical BAF), and polybromo-associated BAF (PBAF). The biological roles for the different enzyme sub-families are poorly described. We knocked down the expression of genes encoding unique subunit proteins for each sub-family, Baf250A, Brd9, and Baf180, which mark the BAF, ncBAF, and PBAF sub-families, respectively, and examined the requirement for each in myoblast differentiation. We found that Baf250A and the BAF complex were required to drive lineage-specific gene expression. KD of Brd9 delayed differentiation. However, while the Baf250A-dependent gene expression profile included myogenic genes, the Brd9-dependent gene expression profile did not, suggesting Brd9 and the ncBAF complex indirectly contributed to differentiation. Baf180 was dispensable for myoblast differentiation. The results distinguish between the roles of the mSWI/SNF enzyme sub-families during myoblast differentiation.
Collapse
Affiliation(s)
- Teresita Padilla-Benavides
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT 06459, USA; (M.O.-F.); (M.D.Z.-E.); (K.Z.)
| | - Monserrat Olea-Flores
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT 06459, USA; (M.O.-F.); (M.D.Z.-E.); (K.Z.)
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; (T.S.); (S.A.S.); (H.W.)
| | - Tapan Sharma
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; (T.S.); (S.A.S.); (H.W.)
| | - Sabriya A. Syed
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; (T.S.); (S.A.S.); (H.W.)
| | - Hanna Witwicka
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; (T.S.); (S.A.S.); (H.W.)
| | - Miriam D. Zuñiga-Eulogio
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT 06459, USA; (M.O.-F.); (M.D.Z.-E.); (K.Z.)
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39086, GRO, Mexico;
| | - Kexin Zhang
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT 06459, USA; (M.O.-F.); (M.D.Z.-E.); (K.Z.)
| | - Napoleon Navarro-Tito
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39086, GRO, Mexico;
| | - Anthony N. Imbalzano
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; (T.S.); (S.A.S.); (H.W.)
| |
Collapse
|
28
|
Katagiri T, Tsukamoto S, Kuratani M, Tsuji S, Nakamura K, Ohte S, Kawaguchi Y, Takaishi K. A blocking monoclonal antibody reveals dimerization of intracellular domains of ALK2 associated with genetic disorders. Nat Commun 2023; 14:2960. [PMID: 37231012 PMCID: PMC10212922 DOI: 10.1038/s41467-023-38746-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 05/15/2023] [Indexed: 05/27/2023] Open
Abstract
Mutations in activin receptor-like kinase 2 (ALK2) can cause the pathological osteogenic signaling seen in some patients with fibrodysplasia ossificans progressiva and other conditions such as diffuse intrinsic pontine glioma. Here, we report that intracellular domain of wild-type ALK2 readily dimerizes in response to BMP7 binding to drive osteogenic signaling. This osteogenic signaling is pathologically triggered by heterotetramers of type II receptor kinases and ALK2 mutant forms, which form intracellular domain dimers in response to activin A binding. We develop a blocking monoclonal antibody, Rm0443, that can suppress ALK2 signaling. We solve the crystal structure of the ALK2 extracellular domain complex with a Fab fragment of Rm0443 and show that Rm0443 induces dimerization of ALK2 extracellular domains in a back-to-back orientation on the cell membrane by binding the residues H64 and F63 on opposite faces of the ligand-binding site. Rm0443 could prevent heterotopic ossification in a mouse model of fibrodysplasia ossificans progressiva that carries the human R206H pathogenic mutant.
Collapse
Affiliation(s)
- Takenobu Katagiri
- Division of Biomedical Sciences, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan.
- Project of Clinical and Basic Research for FOP, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan.
| | - Sho Tsukamoto
- Division of Biomedical Sciences, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan
- Project of Clinical and Basic Research for FOP, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan
| | - Mai Kuratani
- Division of Biomedical Sciences, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan
| | - Shinnosuke Tsuji
- Specialty Medicine Research Laboratories I, R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Kensuke Nakamura
- Modality Research Laboratories, Biologics Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Satoshi Ohte
- Division of Biomedical Sciences, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan
- Graduate School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Yoshiro Kawaguchi
- Modality Research Laboratories, Biologics Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Kiyosumi Takaishi
- Specialty Medicine Research Laboratories I, R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| |
Collapse
|
29
|
Katayama T, Chigi Y, Okamura D. The ensured proliferative capacity of myoblast in serum-reduced conditions with Methyl-β-cyclodextrin. Front Cell Dev Biol 2023; 11:1193634. [PMID: 37250904 PMCID: PMC10213241 DOI: 10.3389/fcell.2023.1193634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
To produce muscle fibers for cultured meat on a large scale, it is important to expand myoblasts in a serum-reduced or serum-free medium to avoid cost, ethical, and environmental issues. Myoblasts such as C2C12 cells differentiate quickly into myotubes and lose their ability to proliferate when the serum-rich medium is replaced with a serum-reduced medium. This study demonstrates that Methyl-β-cyclodextrin (MβCD), a starch-derived agent that depletes cholesterol, can inhibit further differentiation of myoblasts at the MyoD-positive stage by reducing plasma membrane cholesterol on C2C12 cells and primary cultured chick muscle cells. Furthermore, MβCD efficiently blocks cholesterol-dependent apoptotic cell death of myoblasts, which is one of the mechanisms by which it inhibits the differentiation of C2C12 myoblast cells, as dead cells of myoblast are necessary for the fusion of adjacent myoblasts during the differentiation process into myotubes. Importantly, MβCD maintains the proliferative capacity of myoblasts only under differentiation conditions with a serum-reduced medium, suggesting that its mitogenic effect is due to its inhibitory effect on myoblast differentiation into myotube. In conclusion, this study provides significant insights into ensuring the proliferative capacity of myoblasts in a future serum-free condition for cultured meat production.
Collapse
Affiliation(s)
- Tomoka Katayama
- Department of Advanced Bioscience, Faculty of Agriculture, Kindai University, Nara, Japan
| | - Yuta Chigi
- Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Daiji Okamura
- Department of Advanced Bioscience, Faculty of Agriculture, Kindai University, Nara, Japan
| |
Collapse
|
30
|
Amemiya H, Yamamoto M, Higa K, Watanabe G, Taniguchi S, Kitamura K, Jeong J, Yanagisawa N, Fukuda KI, Abe S. Effects of Myostatin on Nuclear Morphology at the Myotendinous Junction. Int J Mol Sci 2023; 24:ijms24076634. [PMID: 37047606 PMCID: PMC10094852 DOI: 10.3390/ijms24076634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/22/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023] Open
Abstract
Myostatin (Myo) is known to suppress skeletal muscle growth, and was recently reported to control tendon homeostasis. The purpose of the present study was to investigate the regulatory involvement of Myo in the myotendinous junction (MTJ) in vivo and in vitro. After Achilles tendon injury in mice, we identified unexpected cell accumulation on the tendon side of the MTJ. At postoperative day 7 (POD7), the nuclei had an egg-like profile, whereas at POD28 they were spindle-shaped. The aspect ratio of nuclei on the tendon side of the MTJ differed significantly between POD7 and POD28 (p = 4.67 × 10−34). We then investigated Myo expression in the injured Achilles tendon. At the MTJ, Myo expression was significantly increased at POD28 relative to POD7 (p = 0.0309). To investigate the action of Myo in vitro, we then prepared laminated sheets of myoblasts (C2C12) and fibroblasts (NIH3T3) (a pseudo MTJ model). Myo did not affect the expression of Pax7 and desmin (markers of muscle development), scleraxis and temonodulin (markers of tendon development), or Sox9 (a common marker of muscle and tendon development) in the cell sheets. However, Myo changed the nuclear morphology of scleraxis-positive cells arrayed at the boundary between the myoblast sheet and the fibroblast sheet (aspect ratio of the cell nuclei, myostatin(+) vs. myostatin(-): p = 0.000134). Myo may strengthen the connection at the MTJ in the initial stages of growth and wound healing.
Collapse
Affiliation(s)
- Hikari Amemiya
- Division of Special Needs Dentistry and Orofacial Pain, Department of Oral Health and Clinical Science, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Masahito Yamamoto
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Kazunari Higa
- Ophthalmology/Cornea Center, Tokyo Dental College Ichikawa General Hospital, 5-11-13 Sugano, Ichikawa, Chiba 272-8513, Japan
| | - Genji Watanabe
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Shuichiro Taniguchi
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Kei Kitamura
- Department of Histology and Developmental Biology, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Juhee Jeong
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, 345 E. 24th Street, New York, NY 10010, USA
| | - Nobuaki Yanagisawa
- Division of Oral Health Sciences, Department of Health Sciences, School of Health and Social Services, Saitama Prefectural University, 820 Sannomia, Koshigaya-shi, Saitama 343-0036, Japan
| | - Ken-ichi Fukuda
- Division of Special Needs Dentistry and Orofacial Pain, Department of Oral Health and Clinical Science, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Shinichi Abe
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| |
Collapse
|
31
|
Autocatalytic base editing for RNA-responsive translational control. Nat Commun 2023; 14:1339. [PMID: 36906659 PMCID: PMC10008589 DOI: 10.1038/s41467-023-36851-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 02/20/2023] [Indexed: 03/13/2023] Open
Abstract
Genetic circuits that control transgene expression in response to pre-defined transcriptional cues would enable the development of smart therapeutics. To this end, here we engineer programmable single-transcript RNA sensors in which adenosine deaminases acting on RNA (ADARs) autocatalytically convert target hybridization into a translational output. Dubbed DART VADAR (Detection and Amplification of RNA Triggers via ADAR), our system amplifies the signal from editing by endogenous ADAR through a positive feedback loop. Amplification is mediated by the expression of a hyperactive, minimal ADAR variant and its recruitment to the edit site via an orthogonal RNA targeting mechanism. This topology confers high dynamic range, low background, minimal off-target effects, and a small genetic footprint. We leverage DART VADAR to detect single nucleotide polymorphisms and modulate translation in response to endogenous transcript levels in mammalian cells.
Collapse
|
32
|
Bigorra Mir M, Charlebois E, Tsyplenkova S, Fillebeen C, Pantopoulos K. Cardiac Hamp mRNA Is Predominantly Expressed in the Right Atrium and Does Not Respond to Iron. Int J Mol Sci 2023; 24:ijms24065163. [PMID: 36982241 PMCID: PMC10049151 DOI: 10.3390/ijms24065163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/10/2023] Open
Abstract
Hepcidin is a liver-derived hormone that controls systemic iron traffic. It is also expressed in the heart, where it acts locally. We utilized cell and mouse models to study the regulation, expression, and function of cardiac hepcidin. Hepcidin-encoding Hamp mRNA was induced upon differentiation of C2C12 cells to a cardiomyocyte-like phenotype and was not further stimulated by BMP6, BMP2, or IL-6, the major inducers of hepatic hepcidin. The mRNAs encoding hepcidin and its upstream regulator hemojuvelin (Hjv) are primarily expressed in the atria of the heart, with ~20-fold higher Hamp mRNA levels in the right vs. left atrium and negligible expression in the ventricles and apex. Hjv−/− mice, a model of hemochromatosis due to suppression of liver hepcidin, exhibit only modest cardiac Hamp deficiency and minor cardiac dysfunction. Dietary iron manipulations did not significantly affect cardiac Hamp mRNA in the atria of wild-type or Hjv−/− mice. Two weeks following myocardial infarction, Hamp was robustly induced in the liver and heart apex but not atria, possibly in response to inflammation. We conclude that cardiac Hamp is predominantly expressed in the right atrium and is partially regulated by Hjv; however, it does not respond to iron and other inducers of hepatic hepcidin.
Collapse
Affiliation(s)
- Maria Bigorra Mir
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Edouard Charlebois
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Sofiya Tsyplenkova
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Carine Fillebeen
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Kostas Pantopoulos
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
- Correspondence: ; Tel.: +1-514-340-8260 (ext. 25293)
| |
Collapse
|
33
|
Ultralong hydroxyapatite nanowires-incorporated dipeptide hydrogel with enhanced mechanical strength and superior in vivo osteogenesis activity. Colloids Surf A Physicochem Eng Asp 2023. [DOI: 10.1016/j.colsurfa.2023.131153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
|
34
|
Valat A, Fourel L, Sales A, Machillot P, Bouin AP, Fournier C, Bosc L, Arboléas M, Bourrin-Reynard I, Wagoner Johnson AJ, Bruckert F, Albigès-Rizo C, Picart C. Interplay between integrins and cadherins to control bone differentiation upon BMP-2 stimulation. Front Cell Dev Biol 2023; 10:1027334. [PMID: 36684447 PMCID: PMC9846056 DOI: 10.3389/fcell.2022.1027334] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 12/05/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction: Upon BMP-2 stimulation, the osteoblastic lineage commitment in C2C12 myoblasts is associated with a microenvironmental change that occurs over several days. How does BMP-2 operate a switch in adhesive machinery to adapt to the new microenvironment and to drive bone cell fate is not well understood. Here, we addressed this question for BMP-2 delivered either in solution or physically bound of a biomimetic film, to mimic its presentation to cells via the extracellular matrix (ECM). Methods: Biommetics films were prepared using a recently developed automated method that enable high content studies of cellular processes. Comparative gene expressions were done using RNA sequencing from the encyclopedia of the regulatory elements (ENCODE). Gene expressions of transcription factors, beta chain (1, 3, 5) integrins and cadherins (M, N, and Cad11) were studied using quantitative PCR. ECM proteins and adhesion receptor expressions were also quantified by Western blots and dot blots. Their spatial organization in and around cells was studied using immuno-stainings. The individual effect of each receptor on osteogenic transcription factors and alkaline phosphatase expression were studied using silencing RNA of each integrin and cadherin receptor. The organization of fibronectin was studied using immuno-staining and quantitative microscopic analysis. Results: Our findings highlight a switch of integrin and cadherin expression during muscle to bone transdifferentiation upon BMP-2 stimulation. This switch occurs no matter the presentation mode, for BMP-2 presented in solution or via the biomimetic film. While C2C12 muscle cells express M-cadherin and Laminin-specific integrins, the BMP-2-induced transdifferentiation into bone cells is associated with an increase in the expression of cadherin-11 and collagen-specific integrins. Biomimetic films presenting matrix-bound BMP-2 enable the revelation of specific roles of the adhesive receptors depending on the transcription factor. Discussion: While β3 integrin and cadherin-11 work in concert to control early pSMAD1,5,9 signaling, β1 integrin and Cadherin-11 control RunX2, ALP activity and fibronectin organization around the cells. In contrast, while β1 integrin is also important for osterix transcriptional activity, Cadherin-11 and β5 integrin act as negative osterix regulators. In addition, β5 integrin negatively regulates RunX2. Our results show that biomimetic films can be used to delinate the specific events associated with BMP-2-mediated muscle to bone transdifferentiation. Our study reveals how integrins and cadherins work together, while exerting distinct functions to drive osteogenic programming. Different sets of integrins and cadherins have complementary mechanical roles during the time window of this transdifferentiation.
Collapse
Affiliation(s)
- Anne Valat
- Grenoble Institute of Engineering, CNRS UMR 5628, LMGP, Grenoble, France
| | - Laure Fourel
- Grenoble Institute of Engineering, CNRS UMR 5628, LMGP, Grenoble, France
| | - Adria Sales
- U1292 Biosanté, INSERM, CEA, CNRS EMR 5000 Biomimetism and Regenerative Medicine, University Grenoble Alpes, Grenoble, France
| | - Paul Machillot
- Grenoble Institute of Engineering, CNRS UMR 5628, LMGP, Grenoble, France
- U1292 Biosanté, INSERM, CEA, CNRS EMR 5000 Biomimetism and Regenerative Medicine, University Grenoble Alpes, Grenoble, France
| | - Anne-Pascale Bouin
- U1209 Institut for Advanced Biosciences, CNRS 5309, University Grenoble Alpes, La Tronche, France
| | - Carole Fournier
- Grenoble Institute of Engineering, CNRS UMR 5628, LMGP, Grenoble, France
| | - Lauriane Bosc
- U1292 Biosanté, INSERM, CEA, CNRS EMR 5000 Biomimetism and Regenerative Medicine, University Grenoble Alpes, Grenoble, France
| | - Mélanie Arboléas
- Grenoble Institute of Engineering, CNRS UMR 5628, LMGP, Grenoble, France
| | - Ingrid Bourrin-Reynard
- U1209 Institut for Advanced Biosciences, CNRS 5309, University Grenoble Alpes, La Tronche, France
| | - Amy J. Wagoner Johnson
- Grenoble Institute of Engineering, CNRS UMR 5628, LMGP, Grenoble, France
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Carle Illinois College of Medicine, Urbana, IL, United States
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, United States
| | - Franz Bruckert
- Grenoble Institute of Engineering, CNRS UMR 5628, LMGP, Grenoble, France
| | - Corinne Albigès-Rizo
- U1209 Institut for Advanced Biosciences, CNRS 5309, University Grenoble Alpes, La Tronche, France
| | - Catherine Picart
- Grenoble Institute of Engineering, CNRS UMR 5628, LMGP, Grenoble, France
- U1292 Biosanté, INSERM, CEA, CNRS EMR 5000 Biomimetism and Regenerative Medicine, University Grenoble Alpes, Grenoble, France
- Institut Universitaire de France (IUF), Paris, France
| |
Collapse
|
35
|
Priddy LB, Krishnan L, Hettiaratchi MH, Karthikeyakannan S, Gupte N, Guldberg RE. Amniotic membrane attenuates heterotopic ossification following high-dose bone morphogenetic protein-2 treatment of segmental bone defects. J Orthop Res 2023; 41:130-140. [PMID: 35340049 PMCID: PMC9512937 DOI: 10.1002/jor.25324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/31/2022] [Accepted: 03/11/2022] [Indexed: 02/04/2023]
Abstract
Treatment of large bone defects with supraphysiological doses of bone morphogenetic protein-2 (BMP-2) has been associated with complications including heterotopic ossification (HO), inflammation, and pain, presumably due to poor spatiotemporal control of BMP-2. We have previously recapitulated extensive HO in our rat femoral segmental defect model by treatment with high-dose BMP-2 (30 μg). Using this model and BMP-2 dose, our objective was to evaluate the utility of a clinically available human amniotic membrane (AM) around the defect space for guided bone regeneration and reduction of HO. We hypothesized that AM surrounding collagen sponge would attenuate heterotopic ossification compared with collagen sponge alone. In vitro, AM retained more BMP-2 than a synthetic poly(ε-caprolactone) membrane through 21 days. In vivo, as hypothesized, the collagen + AM resulted in significantly less heterotopic ossification and correspondingly, lower total bone volume (BV), compared with collagen sponge alone. Although bone formation within the defect was delayed with AM around the defect, by 12 weeks, defect BVs were equivalent. Torsional stiffness was significantly reduced with AM but was equivalent to that of intact bone. Collagen + AM resulted in the formation of dense fibrous tissue and mineralized tissue, while the collagen group contained primarily mineralized tissue surrounded by marrow-like structures. Especially in conjunction with high doses of growth factor delivered via collagen sponge, these findings suggest AM may be effective as an overlay adjacent to bone healing sites to spatially direct bone regeneration and minimize heterotopic ossification.
Collapse
Affiliation(s)
- Lauren B. Priddy
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, GA 30332, USA
- Current affiliation: Department of Agricultural and Biological Engineering, Mississippi State University, 130 Creelman Street, Mississippi State, MS 39762, USA
| | - Laxminarayanan Krishnan
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
| | - Marian H. Hettiaratchi
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, GA 30332, USA
- Current affiliation: Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, 6231 University of Oregon, Eugene, OR 97403, USA
| | - Sukhita Karthikeyakannan
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, GA 30332, USA
| | - Nikhil Gupte
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, GA 30332, USA
| | - Robert E. Guldberg
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive NW, Atlanta, GA 30332, USA
- Current affiliation: Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, 6231 University of Oregon, Eugene, OR 97403, USA
| |
Collapse
|
36
|
Charbe NB, Tambuwala M, Palakurthi SS, Warokar A, Hromić‐Jahjefendić A, Bakshi H, Zacconi F, Mishra V, Khadse S, Aljabali AA, El‐Tanani M, Serrano‐Aroca Ã, Palakurthi S. Biomedical applications of three-dimensional bioprinted craniofacial tissue engineering. Bioeng Transl Med 2023; 8:e10333. [PMID: 36684092 PMCID: PMC9842068 DOI: 10.1002/btm2.10333] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 02/06/2023] Open
Abstract
Anatomical complications of the craniofacial regions often present considerable challenges to the surgical repair or replacement of the damaged tissues. Surgical repair has its own set of limitations, including scarcity of the donor tissues, immune rejection, use of immune suppressors followed by the surgery, and restriction in restoring the natural aesthetic appeal. Rapid advancement in the field of biomaterials, cell biology, and engineering has helped scientists to create cellularized skeletal muscle-like structures. However, the existing method still has limitations in building large, highly vascular tissue with clinical application. With the advance in the three-dimensional (3D) bioprinting technique, scientists and clinicians now can produce the functional implants of skeletal muscles and bones that are more patient-specific with the perfect match to the architecture of their craniofacial defects. Craniofacial tissue regeneration using 3D bioprinting can manage and eliminate the restrictions of the surgical transplant from the donor site. The concept of creating the new functional tissue, exactly mimicking the anatomical and physiological function of the damaged tissue, looks highly attractive. This is crucial to reduce the donor site morbidity and retain the esthetics. 3D bioprinting can integrate all three essential components of tissue engineering, that is, rehabilitation, reconstruction, and regeneration of the lost craniofacial tissues. Such integration essentially helps to develop the patient-specific treatment plans and damage site-driven creation of the functional implants for the craniofacial defects. This article is the bird's eye view on the latest development and application of 3D bioprinting in the regeneration of the skeletal muscle tissues and their application in restoring the functional abilities of the damaged craniofacial tissue. We also discussed current challenges in craniofacial bone vascularization and gave our view on the future direction, including establishing the interactions between tissue-engineered skeletal muscle and the peripheral nervous system.
Collapse
Affiliation(s)
- Nitin Bharat Charbe
- Irma Lerma Rangel College of PharmacyTexas A&M Health Science CenterKingsvilleTexasUSA
| | - Murtaza Tambuwala
- School of Pharmacy and Pharmaceutical ScienceUlster UniversityColeraineUK
| | | | - Amol Warokar
- Department of PharmacyDadasaheb Balpande College of PharmacyNagpurIndia
| | - Altijana Hromić‐Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural SciencesInternational University of SarajevoSarajevoBosnia and Herzegovina
| | - Hamid Bakshi
- School of Pharmacy and Pharmaceutical ScienceUlster UniversityColeraineUK
| | - Flavia Zacconi
- Departamento de Quimica Orgánica, Facultad de Química y de FarmaciaPontificia Universidad Católica de ChileSantiagoChile
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological SciencesPontificia Universidad Católica de ChileSantiagoChile
| | - Vijay Mishra
- School of Pharmaceutical SciencesLovely Professional UniversityPhagwaraIndia
| | - Saurabh Khadse
- Department of Pharmaceutical ChemistryR.C. Patel Institute of Pharmaceutical Education and ResearchDhuleIndia
| | - Alaa A. Aljabali
- Faculty of Pharmacy, Department of Pharmaceutical SciencesYarmouk UniversityIrbidJordan
| | - Mohamed El‐Tanani
- Pharmacological and Diagnostic Research Centre, Faculty of PharmacyAl‐Ahliyya Amman UniversityAmmanJordan
| | - Ãngel Serrano‐Aroca
- Biomaterials and Bioengineering Lab Translational Research Centre San Alberto MagnoCatholic University of Valencia San Vicente MártirValenciaSpain
| | - Srinath Palakurthi
- Irma Lerma Rangel College of PharmacyTexas A&M Health Science CenterKingsvilleTexasUSA
| |
Collapse
|
37
|
MiRNAs and snoRNAs in Bone Metastasis: Functional Roles and Clinical Potential. Cancers (Basel) 2022; 15:cancers15010242. [PMID: 36612237 PMCID: PMC9818347 DOI: 10.3390/cancers15010242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/19/2022] [Accepted: 12/26/2022] [Indexed: 01/03/2023] Open
Abstract
Bone is a frequent site of metastasis. Bone metastasis is associated with a short-term prognosis in cancer patients, and current treatments aim to slow its growth, but are rarely curative. Thus, revealing molecular mechanisms that explain why metastatic cells are attracted to the bone micro-environment, and how they successfully settle in the bone marrow-taking advantage over bone resident cells-and grow into macro-metastasis, is essential to propose new therapeutic approaches. MicroRNAs and snoRNAs are two classes of small non-coding RNAs that post-transcriptionally regulate gene expression. Recently, microRNAs and snoRNAs have been pointed out as important players in bone metastasis by (i) preparing the pre-metastatic niche, directly and indirectly affecting the activities of osteoclasts and osteoblasts, (ii) promoting metastatic properties within cancer cells, and (iii) acting as mediators within cells to support cancer cell growth in bone. This review aims to highlight the importance of microRNAs and snoRNAs in metastasis, specifically in bone, and how their roles can be linked together. We then discuss how microRNAs and snoRNAs are secreted by cancer cells and be found as extracellular vesicle cargo. Finally, we provide evidence of how microRNAs and snoRNAs can be potential therapeutic targets, at least in pre-clinical settings, and how their detection in liquid biopsies can be a useful diagnostic and/or prognostic biomarker to predict the risk of relapse in cancer patients.
Collapse
|
38
|
Kubatzky KF. Pasteurella multocida toxin - lessons learned from a mitogenic toxin. Front Immunol 2022; 13:1058905. [PMID: 36591313 PMCID: PMC9800868 DOI: 10.3389/fimmu.2022.1058905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
The gram-negative, zoonotic bacterium Pasteurella multocida was discovered in 1880 and found to be the causative pathogen of fowl cholera. Pasteurella-related diseases can be found in domestic and wild life animals such as buffalo, sheep, goat, deer and antelope, cats, dogs and tigers and cause hemorrhagic septicemia in cattle, rhinitis or pneumonia in rabbits or fowl cholera in poultry and birds. Pasteurella multocida does not play a major role in the immune-competent human host, but can be found after animal bites or in people with close contact to animals. Toxigenic strains are most commonly found in pigs and express a phage-encoded 146 kDa protein, the Pasteurella multocida toxin (PMT). Toxin-expressing strains cause atrophic rhinitis where nasal turbinate bones are destroyed through the inhibition of bone building osteoblasts and the activation of bone resorbing osteoclasts. After its uptake through receptor-mediated endocytosis, PMT specifically targets the alpha subunit of several heterotrimeric G proteins and constitutively activates them through deamidation of a glutamine residue to glutamate in the alpha subunit. This results in cytoskeletal rearrangement, proliferation, differentiation and survival of cells. Because of the toxin's mitogenic effects, it was suggested that it might have carcinogenic properties, however, no link between Pasteurella infections and cell transformation could be established, neither in tissue culture models nor through epidemiological data. In the recent years it was shown that the toxin not only affects bone, but also the heart as well as basically all cells of innate and adaptive immunity. During the last decade the focus of research shifted from signal transduction processes to understanding how the bacteria might benefit from a bone-destroying toxin. The primary function of PMT seems to be the modulation of immune cell activation which at the same time creates an environment permissive for osteoclast formation. While the disease is restricted to pigs, the implications of the findings from PMT research can be used to explore human diseases and have a high translational potential. In this review our current knowledge will be summarized and it will be discussed what can be learned from using PMT as a tool to understand human pathologies.
Collapse
Affiliation(s)
- Katharina F. Kubatzky
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
39
|
Identification of Agents That Ameliorate Hyperphosphatemia-Suppressed Myogenin Expression Involved in the Nrf2/p62 Pathway in C2C12 Skeletal Muscle Cells. Int J Mol Sci 2022; 23:ijms232315324. [PMID: 36499650 PMCID: PMC9736935 DOI: 10.3390/ijms232315324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/06/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Hyperphosphatemia can occur as a result of reduced phosphate (Pi) excretion in cases of kidney dysfunction, which can induce muscle wasting and suppress myogenic differentiation. Higher Pi suppresses myogenic differentiation and promotes muscle atrophy through canonical (oxidative stress-mediated) and noncanonical (p62-mediated) activation of nuclear factor erythroid 2-related factor 2 (Nrf2) signaling. However, the crosstalk between myogenin and Nrf2/p62 and potential drug(s) for the regulation of myogenin expression needed to be addressed. In this study, we further identified that myogenin may negatively regulate Nrf2 and p62 protein levels in the mouse C2C12 muscle cell line. In the drug screening analysis, we identified N-acetylcysteine, metformin, phenformin, berberine, 4-chloro-3-ethylphenol, cilostazol, and cilomilast as ameliorating the induction of Nrf2 and p62 expression and reduction in myogenin expression that occur due to high Pi. We further elucidated that doxorubicin and hydrogen peroxide reduced the amount of myogenin protein mediated through the Kelch-like ECH-associated protein 1/Nrf2 pathway, differently from the mechanism of high Pi. The dual functional roles of L-ascorbic acid (L-AA) were found to be dependent on the working concentration, where concentrations below 1 mM L-AA reversed the effect of high Pi on myogenin and those above 1 mM L-AA had a similar effect of high Pi on myogenin when used alone. L-AA exacerbated the effect of hydrogen peroxide on myogenin protein and had no further effect of doxorubicin on myogenin protein. In summary, our results further our understanding of the crosstalk between myogenin and Nrf2, with the identification and verification of several potential drugs that can be applied in rescuing the decline of myogenin due to high Pi in muscle cells.
Collapse
|
40
|
Wang B, Zhao Q, Gong X, Wang C, Bai Y, Wang H, Zhou J, Rong X. Transmembrane anterior posterior transformation 1 regulates BMP signaling and modulates the protein stability of SMAD1/5. J Biol Chem 2022; 298:102684. [PMID: 36370851 PMCID: PMC9763856 DOI: 10.1016/j.jbc.2022.102684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 10/26/2022] [Accepted: 11/01/2022] [Indexed: 11/11/2022] Open
Abstract
The bone morphogenetic protein (BMP) signaling pathway plays pivotal roles in various biological processes during embryogenesis and adult homeostasis. Transmembrane anterior posterior transformation 1 (TAPT1) is an evolutionarily conserved protein involved in murine axial skeletal patterning. Genetic defects in TAPT1 result in complex lethal osteochondrodysplasia. However, the specific cellular activity of TAPT1 is not clear. Herein, we report that TAPT1 inhibits BMP signaling and destabilizes the SMAD1/5 protein by facilitating its interaction with SMURF1 E3 ubiquitin ligase, which leads to SMAD1/5 proteasomal degradation. In addition, we found that the activation of BMP signaling facilitates the redistribution of TAPT1 and promotes its association with SMAD1. TAPT1-deficient murine C2C12 myoblasts or C3H/10T1/2 mesenchymal stem cells exhibit elevated SMAD1/5/9 protein levels, which amplifies BMP activation, in turn leading to a boost in the transdifferentiation or differentiation processing of these distinct TAPT1-deficient cell lines changing into mature osteoblasts. Furthermore, the enhancing effect of TAPT1 deficiency on osteogenic differentiation of C3H/10T1/2 cells was observed in an in vivo ectopic bone formation model. Importantly, a subset of TAPT1 mutations identified in humans with lethal skeletal dysplasia exhibited gain-of-function activity on SMAD1 protein levels. Thus, this finding elucidates the role of TAPT1 in the regulation of SMAD1/5 protein stability for controlling BMP signaling.
Collapse
Affiliation(s)
- Bo Wang
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China
| | - Qian Zhao
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China
| | - Xiaoxia Gong
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China
| | - Caixia Wang
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China
| | - Yan Bai
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China
| | - Hongying Wang
- Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, Key Laboratory of State Ethnic Affairs Commission for Biological Technology, College of Life Sciences, South-Central Minzu University, Wuhan, China
| | - Jianfeng Zhou
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China.
| | - Xiaozhi Rong
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China.
| |
Collapse
|
41
|
Hao L, Wang A, Fu J, Sen Liang, Han Q, Jing Y, Li J, Li Q, Bai S, Seeberger PH, Yin J. Biomineralized Dipeptide Self-Assembled Hydrogel with Ultrahigh Mechanical Strength and Osteoinductivity for Bone Regeneration. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.130622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
42
|
Tholen P, Brown CN, Keil C, Bayir A, Zeng HH, Haase H, Thompson RB, Lengyel I, Yücesan G. A 2,7-dichlorofluorescein derivative to monitor microcalcifications. MOLECULAR SYSTEMS DESIGN & ENGINEERING 2022; 7:1415-1421. [PMID: 37927331 PMCID: PMC10624163 DOI: 10.1039/d2me00185c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Herein, we report the crystal structure of 2,7-dichlorofluorescein methyl ester (DCF-ME) and its fluorescence response to hydroxyapatite binding. The reported fluorophore is very selective for staining the bone matrix and provides turn-on fluorescence upon hydroxyapatite binding. The reported fluorophore can readily pass the cell membrane of the C2C12 cell line, and it is non-toxic for the cell line. The reported fluorophore DCF-ME may find applications in monitoring bone remodeling and microcalcification as an early diagnosis tool for breast cancer and age-related macular degeneration.
Collapse
Affiliation(s)
- Patrik Tholen
- Institute for Food Chemistry and Toxicology, Germany, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Connor N Brown
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Claudia Keil
- Institute for Food Chemistry and Toxicology, Germany, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Ali Bayir
- The Department of Chemistry, Yıldız Technical University, 34220, Esenler, Istanbul, Turkey
| | - Hui-Hui Zeng
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Hajo Haase
- Institute for Food Chemistry and Toxicology, Germany, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Richard B Thompson
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Imre Lengyel
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Gündoğ Yücesan
- Institute for Food Chemistry and Toxicology, Germany, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| |
Collapse
|
43
|
Oliver‐Cervelló L, Martin‐Gómez H, Mandakhbayar N, Jo Y, Cavalcanti‐Adam EA, Kim H, Ginebra M, Lee J, Mas‐Moruno C. Mimicking Bone Extracellular Matrix: From BMP-2-Derived Sequences to Osteogenic-Multifunctional Coatings. Adv Healthc Mater 2022; 11:e2201339. [PMID: 35941083 PMCID: PMC11468143 DOI: 10.1002/adhm.202201339] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Indexed: 01/28/2023]
Abstract
Cell-material interactions are regulated by mimicking bone extracellular matrix on the surface of biomaterials. In this regard, reproducing the extracellular conditions that promote integrin and growth factor (GF) signaling is a major goal to trigger bone regeneration. Thus, the use of synthetic osteogenic domains derived from bone morphogenetic protein 2 (BMP-2) is gaining increasing attention, as this strategy is devoid of the clinical risks associated with this molecule. In this work, the wrist and knuckle epitopes of BMP-2 are screened to identify peptides with potential osteogenic properties. The most active sequences (the DWIVA motif and its cyclic version) are combined with the cell adhesive RGD peptide (linear and cyclic variants), to produce tailor-made biomimetic peptides presenting the bioactive cues in a chemically and geometrically defined manner. Such multifunctional peptides are next used to functionalize titanium surfaces. Biological characterization with mesenchymal stem cells demonstrates the ability of the biointerfaces to synergistically enhance cell adhesion and osteogenic differentiation. Furthermore, in vivo studies in rat calvarial defects prove the capacity of the biomimetic coatings to improve new bone formation and reduce fibrous tissue thickness. These results highlight the potential of mimicking integrin-GF signaling with synthetic peptides, without the need for exogenous GFs.
Collapse
Affiliation(s)
- Lluís Oliver‐Cervelló
- BiomaterialsBiomechanics and Tissue Engineering GroupDepartment of Materials Science and EngineeringUniversitat Politècnica de Catalunya (UPC)Barcelona08019Spain
- Barcelona Research Center in Multiscale Science and EngineeringUPCBarcelona08019Spain
| | - Helena Martin‐Gómez
- BiomaterialsBiomechanics and Tissue Engineering GroupDepartment of Materials Science and EngineeringUniversitat Politècnica de Catalunya (UPC)Barcelona08019Spain
- Barcelona Research Center in Multiscale Science and EngineeringUPCBarcelona08019Spain
| | - Nandin Mandakhbayar
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan330‐714Republic of Korea
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan330‐714Republic of Korea
- Department of Biomaterials ScienceSchool of DentistryDankook UniversityCheonan330‐714Republic of Korea
| | - Young‐Woo Jo
- Neobiotech Co.Ltd R&D CenterSeoul08381Republic of Korea
| | - Elisabetta Ada Cavalcanti‐Adam
- Department of Cellular BiophysicsGrowth Factor Mechanobiology groupMax Planck Institute for Medical Research Jahnstraße 2969120HeidelbergGermany
| | - Hae‐Won Kim
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan330‐714Republic of Korea
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan330‐714Republic of Korea
- Department of Biomaterials ScienceSchool of DentistryDankook UniversityCheonan330‐714Republic of Korea
| | - Maria‐Pau Ginebra
- BiomaterialsBiomechanics and Tissue Engineering GroupDepartment of Materials Science and EngineeringUniversitat Politècnica de Catalunya (UPC)Barcelona08019Spain
- Barcelona Research Center in Multiscale Science and EngineeringUPCBarcelona08019Spain
- Institute for Bioengineering of CataloniaBarcelona08028Spain
| | - Jung‐Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan330‐714Republic of Korea
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan330‐714Republic of Korea
- Department of Biomaterials ScienceSchool of DentistryDankook UniversityCheonan330‐714Republic of Korea
| | - Carlos Mas‐Moruno
- BiomaterialsBiomechanics and Tissue Engineering GroupDepartment of Materials Science and EngineeringUniversitat Politècnica de Catalunya (UPC)Barcelona08019Spain
- Barcelona Research Center in Multiscale Science and EngineeringUPCBarcelona08019Spain
| |
Collapse
|
44
|
Teo YC, Park EJ, Guo J, Abbas A, Smith RAA, Goh D, Yeong JPS, Cool S, Teo P. Bioactive PCL-Peptide and PLA-Peptide Brush Copolymers for Bone Tissue Engineering. ACS APPLIED BIO MATERIALS 2022; 5:4770-4778. [PMID: 36101969 DOI: 10.1021/acsabm.2c00455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We report the modular synthesis of bioactive brush-type polycaprolactone-peptide and polylactide-peptide copolymers for applications in bone tissue engineering. The brush copolymers containing pendant side chains of polycaprolactone (PCL) or polylactide (PLA) and PEGylated peptides, including linear Arg-Gly-Asp and collagen-like peptide (Gly-Pro-Hyp)3, were synthesized by ring-opening metathesis polymerization with high conversions and low dispersities (<1.5). These PCL-peptide and PLA-peptide copolymers exhibited good thermal stability for material processing using melt-extrusion-based methods. The copolymers were blended with commercial PCL or PLA, extruded into filaments, and 3D printed using fused filament fabrication methods. These bioactive PCL and PLA materials promoted osteogenic differentiation in vitro and showed good biocompatibility in in vivo murine model study. The promising results presented herein will serve as a useful guide for the design and functionalization of PCL or PLA materials for use in bone tissue engineering.
Collapse
Affiliation(s)
- Yew Chin Teo
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research, 2 Fusionopolis Way, Innovis, Singapore 138634
| | - Eun Ju Park
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research, 2 Fusionopolis Way, Innovis, Singapore 138634
| | - Jiayi Guo
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research, 2 Fusionopolis Way, Innovis, Singapore 138634
| | - Asyraf Abbas
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research, 2 Fusionopolis Way, Innovis, Singapore 138634
| | - Raymond Alexander Alfred Smith
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Singapore 138673
| | - Denise Goh
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Singapore 138673
| | - Joe Poh Sheng Yeong
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Singapore 138673
| | - Simon Cool
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, 61 Biopolis Drive, Singapore 138673
| | - Peili Teo
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research, 2 Fusionopolis Way, Innovis, Singapore 138634
| |
Collapse
|
45
|
Nagar G, Mittal P, Gupta SRR, Pahuja M, Sanger M, Mishra R, Singh A, Singh IK. Multi-omics therapeutic perspective on ACVR1 gene: from genetic alterations to potential targeting. Brief Funct Genomics 2022; 22:123-142. [PMID: 36003055 DOI: 10.1093/bfgp/elac026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/04/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Activin A receptor type I (ACVR1), a transmembrane serine/threonine kinase, belongs to the transforming growth factor-β superfamily, which signals via phosphorylating the downstream effectors and SMAD transcription factors. Its central role in several biological processes and intracellular signaling is well known. Genetic variation in ACVR1 has been associated with a rare disease, fibrodysplasia ossificans progressive, and its somatic alteration is reported in rare cancer diffuse intrinsic pontine glioma. Furthermore, altered expression or variation of ACVR1 is associated with multiple pathologies such as polycystic ovary syndrome, congenital heart defects, diffuse idiopathic skeletal hyperostosis, posterior fossa ependymoma and other malignancies. Recent advancements have witnessed ACVR1 as a potential pharmacological target, and divergent promising approaches for its therapeutic targeting have been explored. This review highlights the structural and functional characteristics of receptor ACVR1, associated signaling pathways, genetic variants in several diseases and cancers, protein-protein interaction, gene expression, regulatory miRNA prediction and potential therapeutic targeting approaches. The comprehensive knowledge will offer new horizons and insights into future strategies harnessing its therapeutic potential.
Collapse
|
46
|
The Kinesin Gene KIF26B Modulates the Severity of Post-Traumatic Heterotopic Ossification. Int J Mol Sci 2022; 23:ijms23169203. [PMID: 36012474 PMCID: PMC9409126 DOI: 10.3390/ijms23169203] [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: 07/19/2022] [Revised: 08/12/2022] [Accepted: 08/14/2022] [Indexed: 11/17/2022] Open
Abstract
The formation of pathological bone deposits within soft tissues, termed heterotopic ossification (HO), is common after trauma. However, the severity of HO formation varies substantially between individuals, from relatively isolated small bone islands through to extensive soft tissue replacement by bone giving rise to debilitating symptoms. The aim of this study was to identify novel candidate therapeutic molecular targets for severe HO. We conducted a genome-wide scan in men and women with HO of varying severity following hip replacement for osteoarthritis. HO severity was dichotomized as mild or severe, and association analysis was performed with adjustment for age and sex. We next confirmed expression of the gene encoded by the lead signal in human bone and in primary human mesenchymal stem cells. We then examined the effect of gene knockout in a murine model of osseous trans-differentiation, and finally we explored transcription factor phosphorylation in key pathways perturbed by the gene. Ten independent signals were suggestively associated with HO severity, with KIF26B as the lead. We subsequently confirmed KIF26B expression in human bone and upregulation upon BMP2-induced osteogenic differentiation in primary human mesenchymal stem cells, and also in a rat tendo-Achilles model of post-traumatic HO. CRISPR-Cas9 mediated knockout of Kif26b inhibited BMP2-induced Runx2, Sp7/Osterix, Col1A1, Alp, and Bglap/Osteocalcin expression and mineralized nodule formation in a murine myocyte model of osteogenic trans-differentiation. Finally, KIF26B deficiency inhibited ERK MAP kinase activation during osteogenesis, whilst augmenting p38 and SMAD 1/5/8 phosphorylation. Taken together, these data suggest a role for KIF26B in modulating the severity of post-traumatic HO and provide a potential novel avenue for therapeutic translation.
Collapse
|
47
|
KYMASIN UP Natural Product Inhibits Osteoclastogenesis and Improves Osteoblast Activity by Modulating Src and p38 MAPK. Nutrients 2022; 14:nu14153053. [PMID: 35893905 PMCID: PMC9370798 DOI: 10.3390/nu14153053] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 11/17/2022] Open
Abstract
The imbalance in osteoblast (OB)-dependent bone formation in favor of osteoclast (OC)-dependent bone resorption is the main cause of loss of tissue mineral mass during bone remodeling leading to osteoporosis conditions. Thus, the suppression of OC activity together with the improvement in the OB activity has been proposed as an effective therapy for maintaining bone mass during aging. We tested the new dietary product, KYMASIN UP containing standardized Withania somnifera, Silybum marianum and Trigonella foenum-graecum herbal extracts or the single extracts in in vitro models mimicking osteoclastogenesis (i.e., RAW 264.7 cells treated with RANKL, receptor activator of nuclear factor kappa-Β ligand) and OB differentiation (i.e., C2C12 myoblasts treated with BMP2, bone morphogenetic protein 2). We found that the dietary product reduces RANKL-dependent TRAP (tartrate-resistant acid phosphatase)-positive cells (i.e., OCs) formation and TRAP activity, and down-regulates osteoclastogenic markers by reducing Src (non-receptor tyrosine kinase) and p38 MAPK (mitogen-activated protein kinase) activation. Withania somnifera appears as the main extract responsible for the anti-osteoclastogenic effect of the product. Moreover, KYMASIN UP maintains a physiological release of the soluble decoy receptor for RANKL, OPG (osteoprotegerin), in osteoporotic conditions and increases calcium mineralization in C2C12-derived OBs. Interestingly, KYMASIN UP induces differentiation in human primary OB-like cells derived from osteoporotic subjects. Based on our results, KYMASIN UP or Withania somnifera-based dietary supplements might be suggested to reverse the age-related functional decline of bone tissue by re-balancing the activity of OBs and OCs, thus improving the quality of life in the elderly and reducing social and health-care costs.
Collapse
|
48
|
Sefkow-Werner J, Le Pennec J, Machillot P, Ndayishimiye B, Castro-Ramirez E, Lopes J, Licitra C, Wang I, Delon A, Picart C, Migliorini E. Automated Fabrication of Streptavidin-Based Self-assembled Materials for High-Content Analysis of Cellular Response to Growth Factors. ACS APPLIED MATERIALS & INTERFACES 2022; 14:10.1021/acsami.2c08272. [PMID: 35849638 PMCID: PMC7614070 DOI: 10.1021/acsami.2c08272] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The automation of liquid-handling routines offers great potential for fast, reproducible, and labor-reduced biomaterial fabrication but also requires the development of special protocols. Competitive systems demand for a high degree in miniaturization and parallelization while maintaining flexibility regarding the experimental design. Today, there are only a few possibilities for automated fabrication of biomaterials inside multiwell plates. We have previously demonstrated that streptavidin-based biomimetic platforms can be employed to study cellular behaviors on biomimetic surfaces. So far, these self-assembled materials were made by stepwise assembly of the components using manual pipetting. In this work, we introduce for the first time a fully automated and adaptable workflow to functionalize glass-bottom multiwell plates with customized biomimetic platforms deposited in single wells using a liquid-handling robot. We then characterize the cell response using automated image acquisition and subsequent analysis. Furthermore, the molecular surface density of the biomimetic platforms was characterized in situ using fluorescence-based image correlation spectroscopy. These measurements were in agreement with standard ex situ spectroscopic ellipsometry measurements. Due to automation, we could do a proof of concept to study the effect of heparan sulfate on the bioactivity of bone morphogenetic proteins on myoblast cells, using four different bone morphogenetic proteins (BMPs) (2, 4, 6, and 7) in parallel, at five increasing concentrations. Using such an automated self-assembly of biomimetic materials, it may be envisioned to further investigate the role of a large variety of extracellular matrix (ECM) components and growth factors on cell signaling.
Collapse
Affiliation(s)
- Julius Sefkow-Werner
- Univ. Grenoble Alpes, CNRS, Grenoble INP**, LMGP, 38000 Grenoble, France
- Univ. Grenoble Alpes, CEA, INSERM, U1292 Biosanté, CNRS EMR 5000 BRM, 3800, Grenoble, France
| | - Jean Le Pennec
- Univ. Grenoble Alpes, CEA, INSERM, U1292 Biosanté, CNRS EMR 5000 BRM, 3800, Grenoble, France
| | - Paul Machillot
- Univ. Grenoble Alpes, CEA, INSERM, U1292 Biosanté, CNRS EMR 5000 BRM, 3800, Grenoble, France
| | - Bertin Ndayishimiye
- Univ. Grenoble Alpes, CEA, INSERM, U1292 Biosanté, CNRS EMR 5000 BRM, 3800, Grenoble, France
| | - Elaine Castro-Ramirez
- Univ. Grenoble Alpes, CEA, INSERM, U1292 Biosanté, CNRS EMR 5000 BRM, 3800, Grenoble, France
| | - Joao Lopes
- Univ. Grenoble Alpes, CEA, INSERM, U1292 Biosanté, CNRS EMR 5000 BRM, 3800, Grenoble, France
| | | | - Irene Wang
- Univ. Grenoble Alpes, CNRS, LiPhy, Grenoble, France
| | | | - Catherine Picart
- Univ. Grenoble Alpes, CNRS, Grenoble INP**, LMGP, 38000 Grenoble, France
- Univ. Grenoble Alpes, CEA, INSERM, U1292 Biosanté, CNRS EMR 5000 BRM, 3800, Grenoble, France
| | - Elisa Migliorini
- Univ. Grenoble Alpes, CNRS, Grenoble INP**, LMGP, 38000 Grenoble, France
- Univ. Grenoble Alpes, CEA, INSERM, U1292 Biosanté, CNRS EMR 5000 BRM, 3800, Grenoble, France
| |
Collapse
|
49
|
Zhang W, Wu Y, Chen Q, Zhang H, Zhou M, Chen K, Cao C, Guo H, Xu J, Liu H, Lin H, Liu C, Liu R. Statistic Copolymers Working as Growth Factor-Binding Mimics of Fibronectin. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200775. [PMID: 35570405 PMCID: PMC9313494 DOI: 10.1002/advs.202200775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/06/2022] [Indexed: 05/07/2023]
Abstract
Growth factors (GFs) play important roles in biological system and are widely used in tissue regeneration. However, their application is greatly hindered by short in vivo lifetime of GFs. GFs are bound to fibronectin dynamically in the extracellular matrix, which inspired the authors to mimic the GF binding domain of fibronectin and design GF-binding amphiphilic copolymers bearing positive charges. The optimal amino acid polymer can bind to a variety of representative GFs, such as bone morphogenetic protein-2 (BMP-2) and TGF-β1 from the transforming growth factor-β superfamily, PDGF-AA and PDGF-BB from the platelet-derived growth factor family, FGF-10 and FGF-21 from the fibroblast growth factor family, epidermal growth factor from the EGF family and hepatocyte growth factor from the plasminogen-related growth factor family, with binding affinities up to the nanomolar level. 3D scaffolds immobilized with the optimal copolymer enable sustained release of loaded BMP-2 without burst release and significantly enhances the in vivo function of BMP-2 for bone formation. This strategy opens new avenues in designing GF-binding copolymers as synthetic mimics of fibronectin for diverse applications.
Collapse
Affiliation(s)
- Wenjing Zhang
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Yueming Wu
- Key Laboratory for Ultrafine Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistryResearch Center for Biomedical Materials of Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Qi Chen
- Key Laboratory for Ultrafine Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistryResearch Center for Biomedical Materials of Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Haodong Zhang
- Key Laboratory for Ultrafine Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistryResearch Center for Biomedical Materials of Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Min Zhou
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Kang Chen
- Key Laboratory for Ultrafine Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistryResearch Center for Biomedical Materials of Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Chuntao Cao
- Key Laboratory for Ultrafine Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistryResearch Center for Biomedical Materials of Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Han Guo
- Shanghai Synchrotron Radiation Facility (SSRF)Shanghai Advanced Research InstituteChinese Academy of SciencesShanghai201204China
| | - Jianrong Xu
- Academy of Integrative MedicineShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Honglai Liu
- School of Chemistry and Molecular EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Haodong Lin
- Department of Orthopedic SurgeryShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistryResearch Center for Biomedical Materials of Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Runhui Liu
- State Key Laboratory of Bioreactor EngineeringEast China University of Science and TechnologyShanghai200237China
- Key Laboratory for Ultrafine Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistryResearch Center for Biomedical Materials of Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| |
Collapse
|
50
|
Pathophysiology and Emerging Molecular Therapeutic Targets in Heterotopic Ossification. Int J Mol Sci 2022; 23:ijms23136983. [PMID: 35805978 PMCID: PMC9266941 DOI: 10.3390/ijms23136983] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/19/2022] [Accepted: 06/22/2022] [Indexed: 12/23/2022] Open
Abstract
The term heterotopic ossification (HO) describes bone formation in tissues where bone is normally not present. Musculoskeletal trauma induces signalling events that in turn trigger cells, probably of mesenchymal origin, to differentiate into bone. The aetiology of HO includes extremely rare but severe, generalised and fatal monogenic forms of the disease; and as a common complex disorder in response to musculoskeletal, neurological or burn trauma. The resulting bone forms through a combination of endochondral and intramembranous ossification, depending on the aetiology, initiating stimulus and affected tissue. Given the heterogeneity of the disease, many cell types and biological pathways have been studied in efforts to find effective therapeutic strategies for the disorder. Cells of mesenchymal, haematopoietic and neuroectodermal lineages have all been implicated in the pathogenesis of HO, and the emerging dominant signalling pathways are thought to occur through the bone morphogenetic proteins (BMP), mammalian target of rapamycin (mTOR), and retinoic acid receptor pathways. Increased understanding of these disease mechanisms has resulted in the emergence of several novel investigational therapeutic avenues, including palovarotene and other retinoic acid receptor agonists and activin A inhibitors that target both canonical and non-canonical signalling downstream of the BMP type 1 receptor. In this article we aim to illustrate the key cellular and molecular mechanisms involved in the pathogenesis of HO and outline recent advances in emerging molecular therapies to treat and prevent HO that have had early success in the monogenic disease and are currently being explored in the common complex forms of HO.
Collapse
|