1
|
Wan L, Yao X, Pan J, Xiang Z, Fu D, Ye Q, Wu F. Crafting the future of bone regeneration: the promise of supramolecular peptide nanofiber hydrogels. Front Bioeng Biotechnol 2025; 13:1514318. [PMID: 40134775 PMCID: PMC11933111 DOI: 10.3389/fbioe.2025.1514318] [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: 10/22/2024] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
Bone tissue engineering has rapidly emerged as an ideal strategy to replace autologous bone grafts, establishing a comprehensive system centered on biomaterial scaffolds, seeding cells, bioactive factors, and biophysical stimulation, thus paving the way for new horizons in surgical bone regeneration. However, the scarcity of suitable materials poses a significant challenge in replicating the intricate multi-layered structure of natural bone tissue. Supramolecular peptide nanofiber hydrogels (SPNHs) have shown tremendous potential as novel biomaterials due to their excellent biocompatibility, biodegradability, tunable mechanical properties, and multifunctionality. Various supramolecular peptides can assemble into nanofiber hydrogels, while bioactive sequences and factors can be embedded through physical adsorption or covalent binding, endowing the hydrogels with diverse biochemical properties. Finally, this review explored the future challenges and prospects of SPNHs in bone tissue engineering, with the aim of providing insights for further advancements in this field.
Collapse
Affiliation(s)
- Longbiao Wan
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoyue Yao
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiali Pan
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ziyang Xiang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Dongjie Fu
- Department of Stomatology, Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qingsong Ye
- Department of Stomatology, Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Sydney Dental School, The University of Sydney, Camperdown, NSW, Australia
| | - Fei Wu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
2
|
Kang J, Li Q, Liu J, Du L, Liu P, Liu F, Wang Y, Shen X, Luo X, Wang N, Wu R, Song L, Wang J, Liu X. Exploring the cellular and molecular basis of murine cardiac development through spatiotemporal transcriptome sequencing. Gigascience 2025; 14:giaf012. [PMID: 39960664 PMCID: PMC11831923 DOI: 10.1093/gigascience/giaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/08/2024] [Accepted: 01/25/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Spatial transcriptomics is a powerful tool that integrates molecular data with spatial information, thereby facilitating a deeper comprehension of tissue morphology and cellular interactions. In our study, we utilized cutting-edge spatial transcriptome sequencing technology to explore the development of the mouse heart and construct a comprehensive spatiotemporal cell atlas of early murine cardiac development. RESULTS Through the analysis of this atlas, we elucidated the spatial organization of cardiac cellular lineages and their interactions during the developmental process. Notably, we observed dynamic changes in gene expression within fibroblasts and cardiomyocytes. Moreover, we identified critical genes, such as Igf2, H19, and Tcap, as well as transcription factors Tcf12 and Plagl1, which may be associated with the loss of myocardial regeneration ability during early heart development. In addition, we successfully identified marker genes, like Adamts8 and Bmp10, that can distinguish between the left and right atria. CONCLUSION Our study provides novel insights into murine cardiac development and offers a valuable resource for future investigations in the field of heart research, highlighting the significance of spatial transcriptomics in understanding the complex processes of organ development.
Collapse
Affiliation(s)
- Jingmin Kang
- BGI Research, Beijing 102601, China
- BGI Research, Shenzhen 518083, China
| | - Qingsong Li
- BGI Research, Beijing 102601, China
- BGI Research, Shenzhen 518083, China
| | - Jie Liu
- Cardiomyopathy Ward, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, China
| | - Lin Du
- BGI Research, Beijing 102601, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peng Liu
- BGI Research, Beijing 102601, China
| | - Fuyan Liu
- BGI Research, Beijing 102601, China
- BGI Research, Shenzhen 518083, China
| | - Yue Wang
- BGI Research, Beijing 102601, China
- BGI Research, Shenzhen 518083, China
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Xunan Shen
- BGI Research, Beijing 102601, China
- BGI Research, Shenzhen 518083, China
| | | | - Ninghe Wang
- Clin Lab, BGI Genomics, Tianjin 300308, China
| | - Renhua Wu
- Clin Lab, BGI Genomics, Tianjin 300308, China
| | - Lei Song
- Cardiomyopathy Ward, Fuwai Hospital, National Center for Cardiovascular Disease , Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100037, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- National Clinical Research Center of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Jizheng Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Xin Liu
- BGI Research, Beijing 102601, China
- BGI Research, Shenzhen 518083, China
| |
Collapse
|
3
|
Ruan X, Jin X, Sun F, Pi J, Jinghu Y, Lin X, Zhang N, Chen G. IGF signaling pathway in bone and cartilage development, homeostasis, and disease. FASEB J 2024; 38:e70031. [PMID: 39206513 DOI: 10.1096/fj.202401298r] [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: 06/08/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
The skeleton plays a fundamental role in the maintenance of organ function and daily activities. The insulin-like growth factor (IGF) family is a group of polypeptide substances with a pronounced role in osteoblast differentiation, bone development, and metabolism. Disturbance of the IGFs and the IGF signaling pathway is inextricably linked with assorted developmental defects, growth irregularities, and jeopardized skeletal structure. Recent findings have illustrated the significance of the action of the IGF signaling pathway via growth factors and receptors and its interactions with dissimilar signaling pathways (Wnt/β-catenin, BMP, TGF-β, and Hh/PTH signaling pathways) in promoting the growth, survival, and differentiation of osteoblasts. IGF signaling also exhibits profound influences on cartilage and bone development and skeletal homeostasis via versatile cell-cell interactions in an autocrine, paracrine, and endocrine manner systemically and locally. Our review summarizes the role and regulatory function as well as a potentially integrated gene network of the IGF signaling pathway with other signaling pathways in bone and cartilage development and skeletal homeostasis, which in turn provides an enlightening insight into visualizing bright molecular targets to be eligible for designing effective drugs to handle bone diseases and maladies, such as osteoporosis, osteoarthritis, and dwarfism.
Collapse
Affiliation(s)
- Xinyi Ruan
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiuhui Jin
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Fuju Sun
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Jiashun Pi
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yihan Jinghu
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xinyi Lin
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Nenghua Zhang
- Clinical Laboratory, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, China
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
4
|
Gu N, Wang Y, Li L, Sui X, Liu Z. The mechanism of lncRNA MALAT1 targeting the miR-124-3p/IGF2BP1 axis to regulate osteogenic differentiation of periodontal ligament stem cells. Clin Oral Investig 2024; 28:219. [PMID: 38492123 DOI: 10.1007/s00784-024-05616-3] [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: 11/14/2023] [Accepted: 03/10/2024] [Indexed: 03/18/2024]
Abstract
OBJECTIVES This study aimed to investigate the regulatory roles of lncRNA MALAT1, miR-124-3p, and IGF2BP1 in osteogenic differentiation of periodontal ligament stem cells (PDLSCs). MATERIALS AND METHODS We characterized PDLSCs by employing quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analyses to evaluate the expression of key osteogenic markers including ALPL, SPP1, and RUNX2. Manipulation of lncRNA MALAT1 and miR-124-3p expression levels was achieved through transfection techniques. In addition, early osteogenic differentiation was assessed via Alkaline phosphatase (ALP) staining, and mineral deposition was quantified using Alizarin Red S (ARS) staining. Cellular localization of lncRNA MALAT1 was determined through Fluorescence In Situ Hybridization (FISH). To elucidate the intricate regulatory network, we conducted dual-luciferase reporter assays to decipher the binding interactions between lncRNA MALAT1 and miR-124-3P as well as between miR-124-3P and IGF2BP1. RESULTS Overexpression of lncRNA MALAT1 robustly promoted osteogenesis in PDLSCs, while its knockdown significantly inhibited the process. We confirmed the direct interaction between miR-124-3p and lncRNA MALAT1, underscoring its role in impeding osteogenic differentiation. Notably, IGF2BP1 was identified as a direct binding partner of lncRNA MALAT1, highlighting its pivotal role within this intricate network. Moreover, we determined the optimal IGF2BP1 concentration (50 ng/ml) as a potent enhancer of osteogenesis, effectively countering the inhibition induced by si-MALAT1. Furthermore, in vivo experiments utilizing rat calvarial defects provided compelling evidence, solidifying lncRNA MALAT1's crucial role in bone formation. CONCLUSIONS Our study reveals the regulatory network involving lncRNA MALAT1, miR-124-3p, and IGF2BP1 in PDLSCs' osteogenic differentiation. CLINICAL RELEVANCE These findings enhance our understanding of lncRNA-mediated osteogenesis, offering potential therapeutic implications for periodontal tissue regeneration and the treatment of bone defects.
Collapse
Affiliation(s)
- Nan Gu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
- Department of Prosthodontics, Hospital of Stomatology, Jilin University, Qinghua Road No.1500, Changchun, 130021, People's Republic of China
| | - Yao Wang
- Department of Stomatology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Lingfeng Li
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
- Department of Prosthodontics, Hospital of Stomatology, Jilin University, Qinghua Road No.1500, Changchun, 130021, People's Republic of China
| | - Xin Sui
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China
- Department of Prosthodontics, Hospital of Stomatology, Jilin University, Qinghua Road No.1500, Changchun, 130021, People's Republic of China
| | - Zhihui Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Department of Prosthodontics, Hospital of Stomatology, Jilin University, Qinghua Road No.1500, Changchun, 130021, People's Republic of China.
| |
Collapse
|
5
|
Zhang C, Wu S, Chen E, Yu L, Wang J, Wu M. ALX1-transcribed LncRNA AC132217.4 promotes osteogenesis and bone healing via IGF-AKT signaling in mesenchymal stem cells. Cell Mol Life Sci 2022; 79:328. [PMID: 35639207 PMCID: PMC11073114 DOI: 10.1007/s00018-022-04338-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/23/2022] [Accepted: 04/29/2022] [Indexed: 11/03/2022]
Abstract
The osteogenic potential of bone marrow mesenchymal stem cells (BMSCs) is critical for bone formation and regeneration. A high non-/delayed-union rate of fracture healing still occurs in specific populations, implying an urgent need to discover novel targets for promoting osteogenesis and bone regeneration. Long non-coding (lnc)RNAs are emerging regulators of multiple physiological processes, including osteogenesis. Based on differential expression analysis of RNA sequencing data, we found that lncRNA AC132217.4, a 3'UTR-overlapping lncRNA of insulin growth factor 2 (IGF2), was highly induced during osteogenic differentiation of BMSCs. Afterward, both gain-of-function and loss-of-function experiments proved that AC132217.4 promotes osteoblast development from BMSCs. As for its molecular mechanism, we found that AC132217.4 binds with IGF2 mRNA to regulate its expression and downstream AKT activation to control osteoblast maturation and function. Furthermore, we identified two splicing factors, splicing component 35 KDa (SC35) and heterogeneous nuclear ribonucleoprotein A1 (HNRNPA1), which regulate the biogenesis of AC132217.4 at the post-transcriptional level. We also identified a transcription factor, ALX1, which regulates AC132217.7 expression at the transcriptional level to promote osteogenesis. Importantly, in-vivo over-expression of AC132217.4 essentially promotes the bone healing process in a murine tibial drill-hole model. Our study demonstrates that lncRNA AC132217.4 is a novel anabolic regulator of BMSC osteogenesis and could be a plausible therapeutic target for improving bone regeneration.
Collapse
Affiliation(s)
- Cui Zhang
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shali Wu
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Erman Chen
- Department of Orthopedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Luyang Yu
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Jinfu Wang
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, China.
| | - Mengrui Wu
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
6
|
Hao Z, Li H, Wang Y, Hu Y, Chen T, Zhang S, Guo X, Cai L, Li J. Supramolecular Peptide Nanofiber Hydrogels for Bone Tissue Engineering: From Multihierarchical Fabrications to Comprehensive Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103820. [PMID: 35128831 PMCID: PMC9008438 DOI: 10.1002/advs.202103820] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/02/2022] [Indexed: 05/03/2023]
Abstract
Bone tissue engineering is becoming an ideal strategy to replace autologous bone grafts for surgical bone repair, but the multihierarchical complexity of natural bone is still difficult to emulate due to the lack of suitable biomaterials. Supramolecular peptide nanofiber hydrogels (SPNHs) are emerging biomaterials because of their inherent biocompatibility, satisfied biodegradability, high purity, facile functionalization, and tunable mechanical properties. This review initially focuses on the multihierarchical fabrications by SPNHs to emulate natural bony extracellular matrix. Structurally, supramolecular peptides based on distinctive building blocks can assemble into nanofiber hydrogels, which can be used as nanomorphology-mimetic scaffolds for tissue engineering. Biochemically, bioactive motifs and bioactive factors can be covalently tethered or physically absorbed to SPNHs to endow various functions depending on physiological and pharmacological requirements. Mechanically, four strategies are summarized to optimize the biophysical microenvironment of SPNHs for bone regeneration. Furthermore, comprehensive applications about SPNHs for bone tissue engineering are reviewed. The biomaterials can be directly used in the form of injectable hydrogels or composite nanoscaffolds, or they can be used to construct engineered bone grafts by bioprinting or bioreactors. Finally, continuing challenges and outlook are discussed.
Collapse
Affiliation(s)
- Zhuowen Hao
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Hanke Li
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Yi Wang
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Yingkun Hu
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Tianhong Chen
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Shuwei Zhang
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Xiaodong Guo
- Department of OrthopedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyJiefang Road 1277Wuhan430022China
| | - Lin Cai
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Jingfeng Li
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| |
Collapse
|
7
|
Stem Cells and Their Derivatives-Implications for Alveolar Bone Regeneration: A Comprehensive Review. Int J Mol Sci 2021; 22:ijms222111746. [PMID: 34769175 PMCID: PMC8583713 DOI: 10.3390/ijms222111746] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 02/07/2023] Open
Abstract
Oral and craniofacial bone defects caused by congenital disease or trauma are widespread. In the case of severe alveolar bone defect, autologous bone grafting has been considered a “gold standard”; however, the procedure has several disadvantages, including limited supply, resorption, donor site morbidity, deformity, infection, and bone graft rejection. In the last few decades, bone tissue engineering combined with stem cell-based therapy may represent a possible alternative to current bone augmentation techniques. The number of studies investigating different cell-based bone tissue engineering methods to reconstruct alveolar bone damage is rapidly rising. As an interdisciplinary field, bone tissue engineering combines the use of osteogenic cells (stem cells/progenitor cells), bioactive molecules, and biocompatible scaffolds, whereas stem cells play a pivotal role. Therefore, our work highlights the osteogenic potential of various dental tissue-derived stem cells and induced pluripotent stem cells (iPSCs), the progress in differentiation techniques of iPSCs into osteoprogenitor cells, and the efforts that have been made to fabricate the most suitable and biocompatible scaffold material with osteoinductive properties for successful bone graft generation. Moreover, we discuss the application of stem cell-derived exosomes as a compelling new form of “stem-cell free” therapy.
Collapse
|
8
|
Sanjurjo-Rodriguez C, Altaie A, Mastbergen S, Baboolal T, Welting T, Lafeber F, Pandit H, McGonagle D, Jones E. Gene Expression Signatures of Synovial Fluid Multipotent Stromal Cells in Advanced Knee Osteoarthritis and Following Knee Joint Distraction. Front Bioeng Biotechnol 2020; 8:579751. [PMID: 33178674 PMCID: PMC7591809 DOI: 10.3389/fbioe.2020.579751] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/16/2020] [Indexed: 12/16/2022] Open
Abstract
Osteoarthritis (OA) is the most common musculoskeletal disorder. Although joint replacement remains the standard of care for knee OA patients, knee joint distraction (KJD), which works by temporarily off-loading the joint for 6–8 weeks, is becoming a novel joint-sparing alternative for younger OA sufferers. The biological mechanisms behind KJD structural improvements remain poorly understood but likely involve joint-resident regenerative cells including multipotent stromal cells (MSCs). In this study, we hypothesized that KJD leads to beneficial cartilage-anabolic and anti-catabolic changes in joint-resident MSCs and investigated gene expression profiles of synovial fluid (SF) MSCs following KJD as compared with baseline. To obtain further insights into the effects of local biomechanics on MSCs present in late OA joints, SF MSC gene expression was studied in a separate OA arthroplasty cohort and compared with subchondral bone (SB) MSCs from medial (more loaded) and lateral (less loaded) femoral condyles from the same joints. In OA arthroplasty cohort (n = 12 patients), SF MSCs expressed lower levels of ossification- and hypotrophy-related genes [bone sialoprotein (IBSP), parathyroid hormone 1 receptor (PTH1R), and runt-related transcription factor 2 (RUNX2)] than did SB MSCs. Interestingly, SF MSCs expressed 5- to 50-fold higher levels of transcripts for classical extracellular matrix turnover molecules matrix metalloproteinase 1 (MMP1), a disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS5), and tissue inhibitor of metalloproteinase-3 (TIMP3), all (p < 0.05) potentially indicating greater cartilage remodeling ability of OA SF MSCs, compared with SB MSCs. In KJD cohort (n = 9 patients), joint off-loading resulted in sustained, significant increase in SF MSC colonies’ sizes and densities and a notable transcript upregulation of key cartilage core protein aggrecan (ACAN) (weeks 3 and 6), as well as reduction in pro-inflammatory C–C motif chemokine ligand 2 (CCL2) expression (weeks 3 and 6). Additionally, early KJD changes (week 3) were marked by significant increases in MSC chondrogenic commitment markers gremlin 1 (GREM1) and growth differentiation factor 5 (GDF5). In combination, our results reveal distinct transcriptomes on joint-resident MSCs from different biomechanical environments and show that 6-week joint off-loading leads to transcriptional changes in SF MSCs that may be beneficial for cartilage regeneration. Biomechanical factors should be certainly considered in the development of novel MSC-based therapies for OA.
Collapse
Affiliation(s)
- Clara Sanjurjo-Rodriguez
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom.,Physiotherapy, Medicine and Biomedical Sciences department, CIBER-BBN, Institute of Biomedical Research of A Coruña (INIBIC)-Centre of Advanced Scientific Researches (CICA), University of A Coruña, A Coruña, Spain
| | - Ala Altaie
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| | - Simon Mastbergen
- University Medical Center Utrecht, Rheumatology & Clinical Immunology, Regenerative Medicine Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Thomas Baboolal
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| | - Tim Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Floris Lafeber
- University Medical Center Utrecht, Rheumatology & Clinical Immunology, Regenerative Medicine Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Hemant Pandit
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom.,NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds, United Kingdom
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom.,NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds, United Kingdom
| | - Elena Jones
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
9
|
Yu S, Zhao Y, Fang TJ, Ge L. Effect of the Soluble Factors Released by Dental Apical Papilla-Derived Stem Cells on the Osteo/Odontogenic, Angiogenic, and Neurogenic Differentiation of Dental Pulp Cells. Stem Cells Dev 2020; 29:795-805. [PMID: 32178575 DOI: 10.1089/scd.2019.0262] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Shi Yu
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yuming Zhao
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| | - Teng Jiaozi Fang
- Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lihong Ge
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
10
|
Diao S, Yang H, Cao Y, Yang D, Fan Z. IGF2 enhanced the osteo-/dentinogenic and neurogenic differentiation potentials of stem cells from apical papilla. J Oral Rehabil 2019; 47 Suppl 1:55-65. [PMID: 31291686 DOI: 10.1111/joor.12859] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 12/27/2022]
Abstract
OBJECTIVES In dental tissue engineering, niche is important for maintaining stem cell function and regenerating the dental tissues. However, there is limited knowledge for the growth factors in niche to maintain the function of stem cells. In this study, we investigated the effect of IGF2, a growth factor in stem cells from apical papilla (SCAPs) niche, on differentiation and proliferation potentials of SCAPs. MATERIALS AND METHODS Recombinant human IGF2 protein (rhIGF2) was used. Cell counting kit-8 assay, Carboxyfluorescein succinimidyl ester assay, alkaline phosphatase (ALP) activity, Alizarin Red staining, quantitative calcium analysis, immunofluorescence staining and real-time RT-PCR were performed to investigate the cell proliferation and differentiation potentials of SCAPs. And proteomic analysis was used to identify the differential secreted proteins. RESULTS By ALP activity assay, we found that 5 ng/mL rhIGF2 might be the optimal concentration for treatment. Then, Alizarin Red staining, quantitative calcium analysis and osteogenesis-related gene expression results showed that 5 ng/mL rhIGF2 could enhance the osteo-/dentinogenic differentiation potentials in SCAPs. Immunofluorescence staining and real-time RT-PCR results showed that neurogenic markers were significantly induced by 5 ng/mL rhIGF2 in SCAPs. Then, CCK-8 assay and CFSE assay results showed that 5 ng/mL rhIGF2 could enhance the cell proliferation in SCAPs. Furthermore, proteomic analysis showed that IGF2 could induce some secreted proteins which function related to the osteogenesis, neurogenesis and cell proliferation. CONCLUSIONS Our results identified that IGF2 might be the potential mediator in niche to promote SCAP function and dental tissue regeneration.
Collapse
Affiliation(s)
- Shu Diao
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China.,Department of Pediatric dentistry, Capital Medical University School of Stomatology, Beijing, China
| | - Haoqing Yang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Yangyang Cao
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Dongmei Yang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China.,Department of Pediatric dentistry, Capital Medical University School of Stomatology, Beijing, China
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| |
Collapse
|
11
|
Kelly RR, McDonald LT, Jensen NR, Sidles SJ, LaRue AC. Impacts of Psychological Stress on Osteoporosis: Clinical Implications and Treatment Interactions. Front Psychiatry 2019; 10:200. [PMID: 31024360 PMCID: PMC6465575 DOI: 10.3389/fpsyt.2019.00200] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/20/2019] [Indexed: 02/06/2023] Open
Abstract
The significant biochemical and physiological effects of psychological stress are beginning to be recognized as exacerbating common diseases, including osteoporosis. This review discusses the current evidence for psychological stress-associated mental health disorders as risk factors for osteoporosis, the mechanisms that may link these conditions, and potential implications for treatment. Traditional, alternative, and adjunctive therapies are discussed. This review is not intended to provide therapeutic recommendations, but, rather, the goal of this review is to delineate potential interactions of psychological stress and osteoporosis and to highlight potential multi-system implications of pharmacological interventions. Review of the current literature identifies several potentially overlapping mechanistic pathways that may be of interest (e.g., glucocorticoid signaling, insulin-like growth factor signaling, serotonin signaling) for further basic and clinical research. Current literature also supports the potential for cross-effects of therapeutics for osteoporosis and mental health disorders. While studies examining a direct link between osteoporosis and chronic psychological stress are limited, the studies reviewed herein suggest that a multi-factorial, personalized approach should be considered for improved patient outcomes in populations experiencing psychological stress, particularly those at high-risk for development of osteoporosis.
Collapse
Affiliation(s)
- Ryan R Kelly
- Research Services, Ralph H. Johnson VA Medical Center, Charleston, SC, United States.,Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Lindsay T McDonald
- Research Services, Ralph H. Johnson VA Medical Center, Charleston, SC, United States.,Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Nathaniel R Jensen
- Research Services, Ralph H. Johnson VA Medical Center, Charleston, SC, United States.,Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Sara J Sidles
- Research Services, Ralph H. Johnson VA Medical Center, Charleston, SC, United States.,Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Amanda C LaRue
- Research Services, Ralph H. Johnson VA Medical Center, Charleston, SC, United States.,Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
12
|
White KA, Olabisi RM. Spatiotemporal Control Strategies for Bone Formation through Tissue Engineering and Regenerative Medicine Approaches. Adv Healthc Mater 2019; 8:e1801044. [PMID: 30556328 DOI: 10.1002/adhm.201801044] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 11/06/2018] [Indexed: 02/06/2023]
Abstract
Global increases in life expectancy drive increasing demands for bone regeneration. The gold standard for surgical bone repair is autografting, which enjoys excellent clinical outcomes; however, it possesses significant drawbacks including donor site morbidity and limited availability. Although collagen sponges delivered with bone morphogenetic protein, type 2 (BMP2) are a common alternative or supplement, they do not efficiently retain BMP2, necessitating extremely high doses to elicit bone formation. Hence, reports of BMP2 complications are rising, including cancer promotion and ectopic bone formation, the latter inducing complications such as breathing difficulties and neurologic impairments. Thus, efforts to exert spatial control over bone formation are increasing. Several tissue engineering approaches have demonstrated the potential for targeted and controlled bone formation. These approaches include biomaterial scaffolds derived from synthetic sources, e.g., calcium phosphates or polymers; natural sources, e.g., bone or seashell; and immobilized biofactors, e.g., BMP2. Although BMP2 is the only protein clinically approved for use in a surgical device, there are several proteins, small molecules, and growth factors that show promise in tissue engineering applications. This review profiles the tissue engineering advances in achieving control over the location and onset of bone formation (spatiotemporal control) toward avoiding the complications associated with BMP2.
Collapse
Affiliation(s)
- Kristopher A. White
- Department of Chemical and Biochemical Engineering; Rutgers University; 98 Brett Road Piscataway NJ 08854 USA
| | - Ronke M. Olabisi
- Department of Biomedical Engineering; Rutgers University; 599 Taylor Road Piscataway NJ 08854 USA
| |
Collapse
|
13
|
Le TH, Christensen OF, Nielsen B, Sahana G. Genome-wide association study for conformation traits in three Danish pig breeds. Genet Sel Evol 2017; 49:12. [PMID: 28118822 PMCID: PMC5259967 DOI: 10.1186/s12711-017-0289-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 01/12/2017] [Indexed: 02/07/2023] Open
Abstract
Background Selection for sound conformation has been widely used as a primary approach to reduce lameness and leg weakness in pigs. Identification of genomic regions that affect conformation traits would help to improve selection accuracy for these lowly to moderately heritable traits. Our objective was to identify genetic factors that underlie leg and back conformation traits in three Danish pig breeds by performing a genome-wide association study followed by meta-analyses. Methods Data on four conformation traits (front leg, back, hind leg and overall conformation) for three Danish pig breeds (23,898 Landrace, 24,130 Yorkshire and 16,524 Duroc pigs) were used for association analyses. Estimated effects of single nucleotide polymorphisms (SNPs) from single-trait association analyses were combined in two meta-analyses: (1) a within-breed meta-analysis for multiple traits to examine if there are pleiotropic genetic variants within a breed; and (2) an across-breed meta-analysis for a single trait to examine if the same quantitative trait loci (QTL) segregate across breeds. SNP annotation was implemented through Sus scrofa Build 10.2 on Ensembl to search for candidate genes. Results Among the 14, 12 and 13 QTL that were detected in the single-trait association analyses for the three breeds, the most significant SNPs explained 2, 2.3 and 11.4% of genetic variance for back quality in Landrace, overall conformation in Yorkshire and back quality in Duroc, respectively. Several candidate genes for these QTL were also identified, i.e. LRPPRC, WRAP73, VRTN and PPARD likely control conformation traits through the regulation of bone and muscle development, and IGF2BP2, GH1, CCND2 and MSH2 can have an influence through growth-related processes. Meta-analyses not only confirmed many significant SNPs from single-trait analyses with higher significance levels, but also detected several additional associated SNPs and suggested QTL with possible pleiotropic effects. Conclusions Our results imply that conformation traits are complex and may be partly controlled by genes that are involved in bone and skeleton development, muscle and fat metabolism, and growth processes. A reliable list of QTL and candidate genes was provided that can be used in fine-mapping and marker assisted selection to improve conformation traits in pigs. Electronic supplementary material The online version of this article (doi:10.1186/s12711-017-0289-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Thu H Le
- Department of Molecular Biology and Genetics, Center for Quantitative Genetics and Genomics, Aarhus University, Tjele, Denmark. .,Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden.
| | - Ole F Christensen
- Department of Molecular Biology and Genetics, Center for Quantitative Genetics and Genomics, Aarhus University, Tjele, Denmark
| | - Bjarne Nielsen
- SEGES Pig Research Centre, Axeltorv, Copenhagen, Denmark
| | - Goutam Sahana
- Department of Molecular Biology and Genetics, Center for Quantitative Genetics and Genomics, Aarhus University, Tjele, Denmark
| |
Collapse
|
14
|
Abstract
Growth factors are essential orchestrators of the normal bone fracture healing response. For non-union defects, delivery of exogenous growth factors to the injured site significantly improves healing outcomes. However, current clinical methods for scaffold-based growth factor delivery are fairly rudimentary, and there is a need for greater spatial and temporal regulation to increase their in vivo efficacy. Various approaches used to provide spatiotemporal control of growth factor delivery from bone tissue engineering scaffolds include physical entrapment, chemical binding, surface modifications, biomineralization, micro- and nanoparticle encapsulation, and genetically engineered cells. Here, we provide a brief review of these technologies, describing the fundamental mechanisms used to regulate release kinetics. Examples of their use in pre-clinical studies are discussed, and their capacities to provide tunable, growth factor delivery are compared. These advanced scaffold systems have the potential to provide safer, more effective therapies for bone regeneration than the systems currently employed in the clinic.
Collapse
|
15
|
Gao X, Usas A, Lu A, Kozemchak A, Tang Y, Poddar M, Sun X, Cummins JH, Huard J. Cyclooxygenase-2 deficiency impairs muscle-derived stem cell-mediated bone regeneration via cellular autonomous and non-autonomous mechanisms. Hum Mol Genet 2016; 25:3216-3231. [PMID: 27354351 DOI: 10.1093/hmg/ddw172] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 04/26/2016] [Accepted: 05/31/2016] [Indexed: 01/10/2023] Open
Abstract
This study investigated the role of cyclooxygenase-2 (COX-2) expression by donor and host cells in muscle-derived stem cell (MDSC)-mediated bone regeneration utilizing a critical size calvarial defect model. We found that BMP4/green fluorescent protein (GFP)-transduced MDSCs formed significantly less bone in COX-2 knock-out (Cox-2KO) than in COX-2 wild-type (WT) mice. BMP4/GFP-transduced Cox-2KO MDSCs also formed significantly less bone than transduced WT MDSCs when transplanted into calvarial defects created in CD-1 nude mice. The impaired bone regeneration in the Cox-2KO MDSCBMP4/GFP group is associated with downregulation of BMP4-pSMAD1/5 signaling, decreased osteogenic differentiation and lowered proliferation capacity after transplantation, compared with WT MDSCBMP4/GFP cells. The Cox-2KO MDSCBMP4/GFP group demonstrated a reduction in cell survival and direct osteogenic differentiation in vitro These effects were mediated in part by the downregulation of Igf1 and Igf2. In addition, the Cox-2KO MDSCBMP4/GFP cells recruited fewer macrophages than the WT MDSC/BMP4/GFP cells in the early phase after injury. We concluded that the bone regeneration capacity of Cox-2KO MDSCs was impaired because of a reduction in cell proliferation and survival capacities, reduction in osteogenic differentiation and a decrease in the ability of the cells to recruit host cells to the injury site.
Collapse
Affiliation(s)
- Xueqin Gao
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Orthopaedic Surgery, Brown Institute for Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO, USA and
| | - Arvydas Usas
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.,Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, Medical Academy, Kaunas, Lithuania
| | - Aiping Lu
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Orthopaedic Surgery, Brown Institute for Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO, USA and
| | - Adam Kozemchak
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ying Tang
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Minakshi Poddar
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xuying Sun
- Department of Orthopaedic Surgery, Brown Institute for Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - James H Cummins
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Orthopaedic Surgery, Brown Institute for Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO, USA and
| | - Johnny Huard
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA .,Department of Orthopaedic Surgery, Brown Institute for Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO, USA and
| |
Collapse
|
16
|
Insulin-like growth factor-II regulates bone sialoprotein gene transcription. Odontology 2015; 104:271-81. [PMID: 25895664 DOI: 10.1007/s10266-015-0205-6] [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: 02/15/2015] [Accepted: 03/15/2015] [Indexed: 10/23/2022]
Abstract
Insulin-like growth factor-I and -II (IGF-I and IGF-II) have been found in bone extracts of several different species, and IGF-II is the most abundant growth factor stored in bone. Bone sialoprotein (BSP) is a noncollagenous extracellular matrix glycoprotein associated with mineralized connective tissues. In this study, we have investigated the regulation of BSP transcription by IGF-II in rat osteoblast-like ROS17/2.8 cells. IGF-II (50 ng/ml) increased BSP mRNA and protein levels after 6-h stimulation, and enhanced luciferase activities of the constructs pLUC3 (-116 to +60), pLUC4 (-425 to +60), pLUC5 (-801 to +60) and pLUC6 (-938 to +60). Effects of IGF-II were inhibited by tyrosine kinase, extracellular signal-regulated kinase1/2 and phosphatidylinositol 3-kinase inhibitors, and abrogated by 2-bp mutations in cAMP response element (CRE), FGF2 response element (FRE) and homeodomain protein-binding site (HOX). The results of gel shift assays showed that nuclear proteins binding to CRE, FRE and HOX sites were increased by IGF-II (50 ng/ml) at 3 and 6 h. CREB1, phospho-CREB1, c-Fos and c-Jun antibodies disrupted the formation of the CRE-protein complexes. Dlx5 and Runx2 antibodies disrupted the FRE- and HOX-protein complex formations. These studies therefore demonstrated that IGF-II increased BSP transcription by targeting CRE, FRE and HOX elements in the proximal promoter of the rat BSP gene. Moreover, phospho-CREB1, c-Fos, c-Jun, Dlx5 and Runx2 transcription factors appear to be key regulators of IGF-II effects on BSP transcription.
Collapse
|
17
|
Li H, Hui X, Li P, Xu A, Li S, Jin S, Wu D. Expression and efficient purification of tag-cleaved active recombinant human insulin-like growth factor-II from Escherichia coli. BIOTECHNOL BIOPROC E 2015. [DOI: 10.1007/s12257-014-0562-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
18
|
Ding W, Li J, Singh J, Alif R, Vazquez-Padron RI, Gomes SA, Hare JM, Shehadeh LA. miR-30e targets IGF2-regulated osteogenesis in bone marrow-derived mesenchymal stem cells, aortic smooth muscle cells, and ApoE-/- mice. Cardiovasc Res 2015; 106:131-42. [PMID: 25678587 DOI: 10.1093/cvr/cvv030] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIMS Activation of an osteogenic transcriptional program contributes to the initiation of aortic calcification in atherosclerosis. The role of microRNAs in regulating aortic calcification is understudied. We tested the hypothesis that miR-30e regulates an osteogenic program in bone marrow-derived mesenchymal stem cells (MSCs), aortic smooth muscle cells (SMCs), and ApoE(-/-) mice. METHODS AND RESULTS In aortas of wild-type mice, we found that miR-30e is highly expressed in medial SMCs. In aortas of old ApoE(-/-) mice, we found that miR-30e transcripts are down-regulated in an inverse relation to the osteogenic markers Runx2, Opn, and Igf2. In vitro, miR-30e over-expression reduced the proliferation of MSCs and SMCs while increasing adipogenic differentiation of MSCs and smooth muscle differentiation of SMCs. In MSCs and SMCs over-expressing miR-30e, microarrays and qPCR showed repression of an osteogenic gene panel including Igf2. Inhibiting miR-30e in MSCs increased Igf2 transcripts. In SMCs, IGF2 recombinant protein rescued miR-30e-repressed osteogenic differentiation. Luciferase and mutagenesis assays showed binding of miR-30e to a novel and essential site at the 3'UTR of Igf2. In ApoE(-/-) mice, injections of antimiR-30e oligos increased Igf2 expression in the aortas and livers and significantly enhanced OPN protein expression and calcium deposition in aortic valves. CONCLUSION miR-30e represses the osteogenic program in MSCs and SMCs by targeting IGF2 and drives their differentiation into adipogenic or smooth muscle lineage, respectively. Our data suggest that down-regulation of miR-30e in aortas with age and atherosclerosis triggers vascular calcification. The miR-30e pathway plays an important regulatory role in vascular diseases.
Collapse
Affiliation(s)
- Wen Ding
- Department of Molecular and Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Jihe Li
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA Department of Medicine, Division of Cardiology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Jayanti Singh
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA Department of Medicine, Division of Cardiology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Razan Alif
- Department of Biochemistry, University of Miami, Coral Gables, FL 33136, USA
| | - Roberto I Vazquez-Padron
- Department of Surgery, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA Vascular Biology Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Samirah A Gomes
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA Department of Medicine, Division of Cardiology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Lina A Shehadeh
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA Department of Medicine, Division of Cardiology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA Vascular Biology Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
19
|
Wu ATH, Aoki T, Sakoda M, Ohta S, Ichimura S, Ito T, Ushida T, Furukawa KS. Enhancing Osteogenic Differentiation of MC3T3-E1 Cells by Immobilizing Inorganic Polyphosphate onto Hyaluronic Acid Hydrogel. Biomacromolecules 2014; 16:166-73. [DOI: 10.1021/bm501356c] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
| | | | - Megumu Sakoda
- Department
of Applied Bioscience, Kanagawa Institute of Technology, 1030 Shimo-ogino, Atsugi, Kanagawa 243-0292, Japan
| | | | - Shigetoshi Ichimura
- Department
of Applied Bioscience, Kanagawa Institute of Technology, 1030 Shimo-ogino, Atsugi, Kanagawa 243-0292, Japan
| | | | | | | |
Collapse
|
20
|
Xiang R, Lee AMC, Eindorf T, Javadmanesh A, Ghanipoor-Samami M, Gugger M, Fitzsimmons CJ, Kruk ZA, Pitchford WS, Leviton AJ, Thomsen DA, Beckman I, Anderson GI, Burns BM, Rutley DL, Xian CJ, Hiendleder S. Widespread differential maternal and paternal genome effects on fetal bone phenotype at mid-gestation. J Bone Miner Res 2014; 29:2392-404. [PMID: 24753181 DOI: 10.1002/jbmr.2263] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 03/09/2014] [Accepted: 04/03/2014] [Indexed: 11/06/2022]
Abstract
Parent-of-origin-dependent (epi)genetic factors are important determinants of prenatal development that program adult phenotype. However, data on magnitude and specificity of maternal and paternal genome effects on fetal bone are lacking. We used an outbred bovine model to dissect and quantify effects of parental genomes, fetal sex, and nongenetic maternal effects on the fetal skeleton and analyzed phenotypic and molecular relationships between fetal muscle and bone. Analysis of 51 bone morphometric and weight parameters from 72 fetuses recovered at day 153 gestation (54% term) identified six principal components (PC1-6) that explained 80% of the variation in skeletal parameters. Parental genomes accounted for most of the variation in bone wet weight (PC1, 72.1%), limb ossification (PC2, 99.8%), flat bone size (PC4, 99.7%), and axial skeletal growth (PC5, 96.9%). Limb length showed lesser effects of parental genomes (PC3, 40.8%) and a significant nongenetic maternal effect (gestational weight gain, 29%). Fetal sex affected bone wet weight (PC1, p < 0.0001) and limb length (PC3, p < 0.05). Partitioning of variation explained by parental genomes revealed strong maternal genome effects on bone wet weight (74.1%, p < 0.0001) and axial skeletal growth (93.5%, p < 0.001), whereas paternal genome controlled limb ossification (95.1%, p < 0.0001). Histomorphometric data revealed strong maternal genome effects on growth plate height (98.6%, p < 0.0001) and trabecular thickness (85.5%, p < 0.0001) in distal femur. Parental genome effects on fetal bone were mirrored by maternal genome effects on fetal serum 25-hydroxyvitamin D (96.9%, p < 0.001) and paternal genome effects on alkaline phosphatase (90.0%, p < 0.001) and their correlations with maternally controlled bone wet weight and paternally controlled limb ossification, respectively. Bone wet weight and flat bone size correlated positively with muscle weight (r = 0.84 and 0.77, p < 0.0001) and negatively with muscle H19 expression (r = -0.34 and -0.31, p < 0.01). Because imprinted maternally expressed H19 regulates growth factors by miRNA interference, this suggests muscle-bone interaction via epigenetic factors.
Collapse
Affiliation(s)
- Ruidong Xiang
- Robinson Research Institute, The University of Adelaide, Adelaide, Australia; JS Davies Epigenetics and Genetics Group, School of Animal and Veterinary Sciences, Roseworthy Campus, The University of Adelaide, Adelaide, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Lauzon MA, Daviau A, Drevelle O, Marcos B, Faucheux N. Identification of a growth factor mimicking the synergistic effect of fetal bovine serum on BMP-9 cell response. Tissue Eng Part A 2014; 20:2524-35. [PMID: 24593122 DOI: 10.1089/ten.tea.2014.0091] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The bone morphogenetic proteins (BMPs) are potent osteogenic molecules that are used for bone repair in delivery systems and in regenerative medicine. We studied the responses of murine MC3T3-E1 preosteoblasts to doses of recombinant human (rh)BMP-9 with and without fetal bovine serum (FBS). rhBMP-2 was used as a control since it is currently approved by the Food and Drug Administration for bone application. We analyzed the major cell signaling pathways and the expression of osteogenic markers. Without FBS, BMP-9 had a similar effect on MC3T3-E1 preosteoblast differentiation in comparison to BMP-2. In contrast, FBS reduced the EC50 of BMP-9 fourfold to sixfold, as determined by osterix gene expression and alkaline phosphatase (ALP) activity, while it had no influence on EC50 of BMP-2. As suggested by MAPK inhibitor assays, FBS could induce an intracellular signaling environment that favors cell response to BMP-9 by inhibiting ERK1/2 activation and increasing p38 phosphorylation. Finally, IGF-2 (100 ng/mL) could mimic the effect of FBS on BMP-9 cell response in terms of MAPK signaling and ALP activity. Thus, the action of BMP-9 on preosteoblast differentiation can be greatly improved by IGF-2. This finding may well be critical for developing optimal growth factor delivery systems and bone tissue engineering strategies.
Collapse
Affiliation(s)
- Marc-Antoine Lauzon
- 1 Canada Research Chair on Cell-Biomaterial Biohybrid Systems, Université de Sherbrooke , Sherbrooke, Canada
| | | | | | | | | |
Collapse
|
22
|
Ngangan AV, Waring JC, Cooke MT, Mandrycky CJ, McDevitt TC. Soluble factors secreted by differentiating embryonic stem cells stimulate exogenous cell proliferation and migration. Stem Cell Res Ther 2014; 5:26. [PMID: 24564947 PMCID: PMC4055104 DOI: 10.1186/scrt415] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 02/10/2014] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Stem cells are being investigated as catalysts of tissue regeneration to either directly replace or promote cellularity lost as a result of traumatic injury or degenerative disease. In many reports, despite low numbers of stably integrated cells, the transient presence of cells delivered or recruited to sites of tissue remodeling globally benefits functional recovery. Such findings have motivated the need to determine how paracrine factors secreted from transplanted cells may be capable of positively impacting endogenous repair processes and somatic cell responses. METHODS Embryonic stem cells were differentiated as embryoid bodies (EBs) in vitro and media conditioned by EBs were collected at different intervals of time. Gene and protein expression analysis of several different growth factors secreted by EBs were examined by polymerase chain reaction and enzyme-linked immunosorbent assay analysis, respectively, as a function of time. The proliferation and migration of fibroblasts and endothelial cells treated with EB conditioned media was examined compared with unconditioned and growth media controls. RESULTS The expression of several growth factors, including bone morphogenic protein-4, insulin-like growth factors and vascular endothelial growth factor-A, increased during the course of embryonic stem cell (ESC) differentiation as EBs. Conditioned media collected from EBs at different stages of differentiation stimulated proliferation and migration of both fibroblasts and endothelial cells, based on 5-bromo-2'-deoxyuridine incorporation and transwell assays, respectively. CONCLUSIONS Overall, these results demonstrate that differentiating ESCs express increasing amounts of various growth factors over time that altogether are capable of stimulating mitogenic and motogenic activity of exogenous cell populations.
Collapse
|
23
|
Konermann A, Lossdörfer S, Jäger A, Chen Y, Götz W. Autoregulation of insulin-like growth factor 2 and insulin-like growth factor-binding protein 6 in periodontal ligament cells in vitro. Ann Anat 2013; 195:527-32. [DOI: 10.1016/j.aanat.2013.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 10/02/2013] [Accepted: 10/04/2013] [Indexed: 11/26/2022]
|
24
|
Induced chondrogenic differentiation of parthenogenetic murine embryonic stem cells by insulin-like growth factor 2 treatment in a three-dimensional culture environment. Tissue Eng Regen Med 2013. [DOI: 10.1007/s13770-013-1100-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
25
|
Bose S, Roy M, Bandyopadhyay A. Recent advances in bone tissue engineering scaffolds. Trends Biotechnol 2012; 30:546-54. [PMID: 22939815 DOI: 10.1016/j.tibtech.2012.07.005] [Citation(s) in RCA: 1251] [Impact Index Per Article: 96.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Revised: 07/23/2012] [Accepted: 07/23/2012] [Indexed: 12/12/2022]
Abstract
Bone disorders are of significant concern due to increase in the median age of our population. Traditionally, bone grafts have been used to restore damaged bone. Synthetic biomaterials are now being used as bone graft substitutes. These biomaterials were initially selected for structural restoration based on their biomechanical properties. Later scaffolds were engineered to be bioactive or bioresorbable to enhance tissue growth. Now scaffolds are designed to induce bone formation and vascularization. These scaffolds are often porous, made of biodegradable materials that harbor different growth factors, drugs, genes, or stem cells. In this review, we highlight recent advances in bone scaffolds and discuss aspects that still need to be improved.
Collapse
Affiliation(s)
- Susmita Bose
- W.M. Keck Biomedical Materials Research Lab, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164-2920, USA.
| | | | | |
Collapse
|
26
|
Karfeld-Sulzer LS, Weber FE. Biomaterial development for oral and maxillofacial bone regeneration. J Korean Assoc Oral Maxillofac Surg 2012. [DOI: 10.5125/jkaoms.2012.38.5.264] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Affiliation(s)
- Lindsay S. Karfeld-Sulzer
- Oral Biotechnology and Bioengineering, Department of Cranio-Maxillofacial and Oral Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Franz E. Weber
- Oral Biotechnology and Bioengineering, Department of Cranio-Maxillofacial and Oral Surgery, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|