1
|
Li Z, Tian Y. Role of NEL‑like molecule‑1 in osteogenesis/chondrogenesis (Review). Int J Mol Med 2025; 55:5. [PMID: 39450541 PMCID: PMC11537270 DOI: 10.3892/ijmm.2024.5446] [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: 08/09/2024] [Accepted: 10/09/2024] [Indexed: 10/26/2024] Open
Abstract
A dynamic balance exists between osteogenesis and osteoclastogenesis in bone tissue, which can lead to several bone diseases, such as osteoporosis, osteoarthritis, bone necrosis and bone defects, in cases of insufficient osteogenesis or excessive osteoclastogenesis. NEL‑like molecule‑1 (NELL‑1) was first discovered in 1999 as an osteogenic factor that can prevent or treat bone diseases by increasing osteogenic levels. To date, research has identified multiple signaling pathways involved in improving osteogenic levels. Furthermore, to apply NELL‑1 in clinical practice, researchers have optimized its osteogenic effect by combining it with other molecules, changing its molecular structure and performing bone tissue engineering. Currently, research on NELL‑1 is gaining increasing attention. In the near future, it will definitely be applied in clinical practice to eliminate diseases. Thus, the present study provides a comprehensive review of NELL‑1 in enhancing osteogenic levels from the perspectives of the molecular mechanism, interactions with other molecules/cells, molecular‑level changes, applications in bone tissue engineering and its expression in tumors, providing a solid theoretical basis for its clinical application.
Collapse
Affiliation(s)
- Zihan Li
- Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yihao Tian
- Department of Pathology, Beifang Hospital of China Medical University, General Hospital of Northern Theater Command, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
2
|
Culiat C, Soni D, Malkes W, Wienhold M, Zhang LH, Henry E, Dragan M, Kar S, Angeles DM, Eaker S, Biswas R. NELL1 variant protein (NV1) modulates hyper-inflammation, Th-1 mediated immune response, and the HIF-1α hypoxia pathway to promote healing in viral-induced lung injury. Biochem Biophys Res Commun 2025; 744:151198. [PMID: 39706056 DOI: 10.1016/j.bbrc.2024.151198] [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: 10/16/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
Research underscores the urgent need for technological innovations to treat lung tissue damage from viral infections and the lasting impact of COVID-19. Our study demonstrates the effectiveness of recombinant human NV1 protein in promoting a pro-healing extracellular matrix that regulates homeostasis in response to excessive tissue reactions caused by infection and injury. NV1 achieves this by calibrating multiple biological mechanisms, including reducing hyperinflammatory cytokine levels (e.g., IFN-γ, TNF-α, IL-10, and IP-10), enhancing the production of proteins involved in viral inactivation and clearance through endocytosis and phagocytosis (e.g., IL-9, IL-1α), regulating pro-clotting and thrombolytic pathways (e.g., downregulates SERPINE 1 and I-TAC during Th1-mediated inflammation), maintaining cell survival under hypoxic conditions via HIF-1α regulation through the M3K5-JNK-AP-1 and TSC2-mTOR pathways, and promoting blood vessel formation. Our findings reveal NV1 as a potential therapeutic candidate for treating severe lung injuries caused by inflammatory and hypoxic conditions from viral infections and related diseases.
Collapse
Affiliation(s)
| | - Dharmendra Soni
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | | | - Mark Wienhold
- NellOne Therapeutics Inc., Knoxville, TN, 37931, USA
| | | | | | | | | | | | - Shannon Eaker
- NellOne Therapeutics Inc., Knoxville, TN, 37931, USA
| | - Roopa Biswas
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| |
Collapse
|
3
|
Sojan JM, Licini C, Marcheggiani F, Carnevali O, Tiano L, Mattioli-Belmonte M, Maradonna F. Bacillus subtilis Modulated the Expression of Osteogenic Markers in a Human Osteoblast Cell Line. Cells 2023; 12:cells12030364. [PMID: 36766709 PMCID: PMC9913848 DOI: 10.3390/cells12030364] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/20/2023] Open
Abstract
Several in vivo trials have previously demonstrated the beneficial effects of the administration of various probiotic forms on bone health. In this study, we explored the potency of two probiotics, Bacillus subtilis and Lactococcus lactis, alone or in combination with vitamin D (VD), to modulate the transcription of genes involved in the ossification process in a human osteoblast cell line. Genes that mark the "osteoblast proliferation phase", such as RUNX2, TGFB1, and ALPL, "extracellular matrix (ECM) maturation", such as SPP1 and SPARC, as well as "ECM mineralization", such as BGN, BGLAP, and DCN, were all highly expressed in osteoblasts treated with B. subtilis extract. The observed increase in the transcription of the ALPL mRNA was further in agreement with its protein levels as observed by Western blot and immunofluorescence. Therefore, this higher transcription and translation of alkaline phosphatase in osteoblasts treated with the B. subtilis extract, indicated its substantial osteogenic impact on human osteoblasts. Although both the probiotic extracts showed no osteogenic synergy with VD, treatment with B. subtilis alone could increase the ECM mineralization, outperforming the effects of L. lactis and even VD. Furthermore, these results supported the validity of employing probiotic extracts rather than live cells to investigate the effects of probiotics in the in vitro systems.
Collapse
Affiliation(s)
- Jerry Maria Sojan
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - Caterina Licini
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Via Tronto 10/a, 60126 Ancona, Italy
| | - Fabio Marcheggiani
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - Oliana Carnevali
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy
- Correspondence: (O.C.); (F.M.); Tel.: +39-0712204990 (O.C.)
| | - Luca Tiano
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - Monica Mattioli-Belmonte
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Via Tronto 10/a, 60126 Ancona, Italy
| | - Francesca Maradonna
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy
- Correspondence: (O.C.); (F.M.); Tel.: +39-0712204990 (O.C.)
| |
Collapse
|
4
|
Tanjaya J, Ha P, Zhang Y, Wang C, Shah Y, Berthiaume E, Pan HC, Shi J, Kwak J, Wu B, Ting K, Zhang X, Soo C. Genetic and pharmacologic suppression of PPARγ enhances NELL-1-stimulated bone regeneration. Biomaterials 2022; 287:121609. [PMID: 35839586 PMCID: PMC10434299 DOI: 10.1016/j.biomaterials.2022.121609] [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: 05/16/2021] [Revised: 05/15/2022] [Accepted: 05/28/2022] [Indexed: 11/02/2022]
Abstract
Recent investigations into mechanisms behind the development of osteoporosis suggest that suppressing PPARγ-mediated adipogenesis can improve bone formation and bone mineral density. In this study, we investigated a co-treatment strategy to enhance bone formation by combining NELL-1, an osteogenic molecule that has been extensively studied for its potential use as a therapeutic for osteoporosis, with two methods of PPARγ suppression. First, we suppressed PPARγ genetically using lentiviral PPARγ-shRNA in immunocompromised mice for a proof of concept. Second, we used a PPARγ antagonist to suppress PPARγ pharmacologically in immunocompetent senile osteopenic mice for clinical transability. We found that the co-treatment strategy significantly increased bone formation, increased the proliferation stage cell population, decreased late apoptosis of primary mouse BMSCs, and increased osteogenic marker mRNA levels in comparison to the single agent treatment groups. The addition of PPARγ suppression to NELL-1 therapy enhanced NELL-1's effects on bone formation by upregulating anabolic processes without altering NELL-1's inhibitory effects on osteoclastic and adipogenic activities. Our findings suggest that combining PPARγ suppression with therapeutic NELL-1 may be a viable method that can be further developed as a novel strategy to reverse bone loss and decrease marrow adiposity in age-related osteoporosis.
Collapse
Affiliation(s)
- Justine Tanjaya
- Section of Orthodontics, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA, 90025
| | - Pin Ha
- Section of Orthodontics, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA, 90025
| | - Yulong Zhang
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California, Los Angeles, Los Angeles, CA, USA, 90025; Weintraub Center for Reconstructive Biotechnology, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA, 90025
| | - Chenchao Wang
- Section of Orthodontics, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA, 90025
| | - Yash Shah
- Section of Orthodontics, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA, 90025
| | - Emily Berthiaume
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA, 90025
| | - Hsin Chuan Pan
- Section of Orthodontics, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA, 90025
| | - Jiayu Shi
- Section of Orthodontics, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA, 90025
| | - Jinny Kwak
- Section of Orthodontics, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA, 90025
| | - Benjamin Wu
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California, Los Angeles, Los Angeles, CA, USA, 90025; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA, 90025; Weintraub Center for Reconstructive Biotechnology, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA, 90025
| | - Kang Ting
- Forsyth Institute, Harvard University, Cambridge, MA, USA, 02142.
| | - Xinli Zhang
- Section of Orthodontics, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA, 90025.
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA, USA, 90025; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA, 90025; Weintraub Center for Reconstructive Biotechnology, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA, 90025.
| |
Collapse
|
5
|
Liu Z, Liu H, Li D, Ma L, Lu T, Sun H, Zhang Y, Yang H. Evaluation of Biomarkers and Immune Microenvironment of Osteoarthritis: Evidence From Omics Data and Machine Learning. Front Genet 2022; 13:905027. [PMID: 35651940 PMCID: PMC9149375 DOI: 10.3389/fgene.2022.905027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 04/11/2022] [Indexed: 12/21/2022] Open
Abstract
Objectives: This study aimed to identify novel biomarkers for osteoarthritis (OA) and explore potential pathological immune cell infiltration. Methods: We identified differentially expressed genes (DEGs) between OA and normal synovial tissues using the limma package in R, and performed enrichment analyses to understand the functions and enriched pathways of DEGs. Weighted gene co-expression network analysis (WGCNA) and distinct machine-learning algorithms were then used to identify hub modules and candidate biomarkers. We assessed the diagnostic value of the candidate biomarkers using receiver operating characteristic (ROC) analysis. We then used the CIBERSORT algorithm to analyze immune cell infiltration patterns, and the Wilcoxon test to screen out hub immune cells that might affect OA occurrence. Finally, the expression levels of hub biomarkers were confirmed by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). Results: We identified 102 up-regulated genes and 110 down-regulated genes. The functional enrichment analysis results showed that DEGs are enriched mainly in immune response pathways. Combining the results of the algorithms and ROC analysis, we identified GUCA1A and NELL1 as potential diagnostic biomarkers for OA, and validated their diagnosibility using an external dataset. Construction of a TF-mRNA-miRNA network enabled prediction of potential candidate compounds targeting hub biomarkers. Immune cell infiltration analyses revealed the expression of hub biomarkers to be correlated with CD8 T cells, memory B cells, M0/M2 macrophages, resting mast cells and resting dendritic cells. qRT-PCR results showed both GUCA1A and NELL1 were significantly increased in OA samples (p < 0.01). All validations are consistent with the microarray hybridization, indicating that GUCA1A and NELL1 may be involved in the pathogenesis of OA. Conclusion: The findings suggest that GUCA1A and NELL1, closely related to OA occurrence and progression, represent new OA candidate markers, and that immune cell infiltration plays a significant role in the progression of OA.
Collapse
Affiliation(s)
- Zhixin Liu
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, China
| | - Heng Liu
- NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, China
| | - Deqiang Li
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, China
| | - Liang Ma
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, China
| | - Tongxin Lu
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, China
| | - Hao Sun
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, China
| | - Yuankai Zhang
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, China
| | - Hui Yang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
6
|
Xu HZ, Su JS. Restoration of critical defects in the rabbit mandible using osteoblasts and vascular endothelial cells co-cultured with vascular stent-loaded nano-composite scaffolds. J Mech Behav Biomed Mater 2021; 124:104831. [PMID: 34555626 DOI: 10.1016/j.jmbbm.2021.104831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 09/04/2021] [Accepted: 09/07/2021] [Indexed: 01/07/2023]
Abstract
The success of large bone defect repair with tissue engineering technology depends mainly on angiogenesis and osteogenesis. In this study, we prepared poly-caprolactone/nano-hydroxyapatite/beta-calcium phosphate (PCL/nHA/β-TCP) composite scaffolds loaded with poly-(lactic-co-glycolic acid)/nano-hydroxyapatite/collagen/heparin sodium (PLGA/nHA/Col/HS) nanofiber small vascular stent by electrospinning and hot press forming-particle leaching methods. Supramolecular electrostatic self-assembly technology was used to modify the surfaces of small vascular stents to aid in hydrophilicity and anticoagulation. The surfaces of composite scaffolds were modified with an Arg-Gly-Asp (RGD) short peptide by physical adsorption to supply cell adhesion sites. The scaffolds were then combined with rabbit bone marrow-derived osteoblasts (OBs) and rabbit bone marrow-derived vascular endothelial cells (RVECs) to construct large, biologically active vascularized tissue-engineered bone in vitro; this bone was then used to repair critical bone defects in rabbit mandibles. Mechanical and biocompatibility testing results showed that PCL/nHA/β-TCP composite scaffolds loaded with small vascular stents had good surface structure, mechanical properties, biocompatibility, and bone-regeneration induction potential. Twelve weeks after implantation, histological analysis and X-ray scans showed that the use of osteoblasts and vascular endothelial cells co-cultured with PCL/nHA/β-TCP scaffolds was sufficient to repair critical defects in rabbit mandibles.
Collapse
Affiliation(s)
- Hong Zhen Xu
- Department of Prosthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Jian Sheng Su
- Department of Prosthodontics, School & Hospital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, Shanghai, China.
| |
Collapse
|
7
|
An HJ, Ko KR, Baek M, Jeong Y, Lee HH, Kim H, Kim DK, Lee SY, Lee S. Pro-Angiogenic and Osteogenic Effects of Adipose Tissue-Derived Pericytes Synergistically Enhanced by Nel-like Protein-1. Cells 2021; 10:cells10092244. [PMID: 34571892 PMCID: PMC8470876 DOI: 10.3390/cells10092244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/19/2021] [Accepted: 08/24/2021] [Indexed: 11/16/2022] Open
Abstract
An important objective of vascularized tissue regeneration is to develop agents for osteonecrosis. We aimed to identify the pro-angiogenic and osteogenic efficacy of adipose tissue-derived (AD) pericytes combined with Nel-like protein-1 (NELL-1) to investigate the therapeutic effects on osteonecrosis. Tube formation and cell migration were assessed to determine the pro-angiogenic efficacy. Vessel formation was evaluated in vivo using the chorioallantoic membrane assay. A mouse model with a 2.5 mm necrotic bone fragment in the femoral shaft was used as a substitute for osteonecrosis in humans. Bone formation was assessed radiographically (plain radiographs, three-dimensional images, and quantitative analyses), and histomorphometric analyses were performed. To identify factors related to the effects of NELL-1, analysis using microarrays, qRT-PCR, and Western blotting was performed. The results for pro-angiogenic efficacy evaluation identified synergistic effects of pericytes and NELL-1 on tube formation, cell migration, and vessel formation. For osteogenic efficacy analysis, the mouse model for osteonecrosis was treated in combination with pericytes and NELL-1, and the results showed maximum bone formation using radiographic images and quantitative analyses, compared with other treatment groups and showed robust bone and vessel formation using histomorphometric analysis. We identified an association between FGF2 and the effects of NELL-1 using array-based analysis. Thus, combinatorial therapy using AD pericytes and NELL-1 may have potential as a novel treatment for osteonecrosis.
Collapse
Affiliation(s)
- Hyun-Ju An
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-Ro, Seongnam-si 13496, Gyeonggi-do, Korea; (H.-J.A.); (M.B.); (Y.J.); (H.H.L.)
| | - Kyung Rae Ko
- Department of Orthopaedic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Korea;
| | - Minjung Baek
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-Ro, Seongnam-si 13496, Gyeonggi-do, Korea; (H.-J.A.); (M.B.); (Y.J.); (H.H.L.)
| | - Yoonhui Jeong
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-Ro, Seongnam-si 13496, Gyeonggi-do, Korea; (H.-J.A.); (M.B.); (Y.J.); (H.H.L.)
| | - Hyeon Hae Lee
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-Ro, Seongnam-si 13496, Gyeonggi-do, Korea; (H.-J.A.); (M.B.); (Y.J.); (H.H.L.)
| | - Hyungkyung Kim
- Department of Pathology, Kyung Hee University Hospital at Gangdong, Kyung Hee University, College of Medicine, 892 Dongnam-Ro, Gangdong-gu, Seoul 05278, Korea;
| | - Do Kyung Kim
- CHA Graduate School of Medicine, 120 Hyeryong-Ro, Pocheon-si 11160, Gyeonggi-do, Korea;
| | - So-Young Lee
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-Ro, Seongnam-si 13496, Gyeonggi-do, Korea;
| | - Soonchul Lee
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-Ro, Seongnam-si 13496, Gyeonggi-do, Korea; (H.-J.A.); (M.B.); (Y.J.); (H.H.L.)
- Correspondence: ; Tel.: +82-317-805-289; Fax: +82-317-083-578
| |
Collapse
|
8
|
Song H, Zhang Y, Zhang Z, Xiong S, Ma X, Li Y. Hydroxyapatite/NELL-1 Nanoparticles Electrospun Fibers for Osteoinduction in Bone Tissue Engineering Application. Int J Nanomedicine 2021; 16:4321-4332. [PMID: 34211273 PMCID: PMC8241815 DOI: 10.2147/ijn.s309567] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/28/2021] [Indexed: 12/27/2022] Open
Abstract
Background As commonly bone defect is a disease of jaw that can seriously affect implant restoration, the bioactive scaffold can be used as potential systems to provide effective repair for bone defect. Purpose A osteoinductive bone tissue engineering scaffold has been prepared in order to explore the effect of bioactive materials on bone tissue engineering. Methods In this study, NELL-1 nanoparticles (Chi/NNP) and nano hydroxyapatite were incorporated in composite scaffolds by electrospinning and characterized using TEM, SEM, contact angle, tensile tests and in vitro drug release. In vitro biological activities such as MC3T3-E1 cell attachment, proliferation and osteogenic activity were studied. Results With the addition of nHA and nanoparticles, the fiber diameter of PCL/BNPs group, PCL/NNPs group and PCL/nHA/NNPs group was significantly increased. Moreover, the hydrophilic hydroxyl group and amino group presented in nHA and nanoparticles had improved the hydrophilicity of the composite fibers. The composite electrospun containing Chi/NNPs can form a double protective barrier which can effectively prolong the release time of NELL-1 growth factor. In addition, the hydroxyapatite/NELL-1 nanoparticles electrospun fibers can promote attachment, proliferation, differentiation of MC3T3-E1 cells and good cytocompatibility, indicating better ability of inducing osteogenic differentiation. Conclusion A multi-functional PCL/nHA/NNPs composite fiber with long-term bioactivity and osteoinductivity was successfully prepared by electrospinning. This potential composite could be used as scaffolds in bone tissue engineering application after in vivo studies.
Collapse
Affiliation(s)
- Hualei Song
- Department of Laboratory, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Yuntao Zhang
- Department of Stomatology, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Zihan Zhang
- Department of Stomatology, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Shijiang Xiong
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Jinan, 250012, People's Republic of China
| | - Xiangrui Ma
- Department of Stomatology, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Yourui Li
- Department of Stomatology, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| |
Collapse
|
9
|
Li C, Mills Z, Zheng Z. Novel cell sources for bone regeneration. MedComm (Beijing) 2021; 2:145-174. [PMID: 34766140 PMCID: PMC8491221 DOI: 10.1002/mco2.51] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/03/2020] [Accepted: 12/09/2020] [Indexed: 01/09/2023] Open
Abstract
A plethora of both acute and chronic conditions, including traumatic, degenerative, malignant, or congenital disorders, commonly induce bone disorders often associated with severe persisting pain and limited mobility. Over 1 million surgical procedures involving bone excision, bone grafting, and fracture repair are performed each year in the U.S. alone, resulting in immense levels of public health challenges and corresponding financial burdens. Unfortunately, the innate self-healing capacity of bone is often inadequate for larger defects over a critical size. Moreover, as direct transplantation of committed osteoblasts is hindered by deficient cell availability, limited cell spreading, and poor survivability, an urgent need for novel cell sources for bone regeneration is concurrent. Thanks to the development in stem cell biology and cell reprogramming technology, many multipotent and pluripotent cells that manifest promising osteogenic potential are considered the regenerative remedy for bone defects. Considering these cells' investigation is still in its relative infancy, each of them offers their own particular challenges that must be conquered before the large-scale clinical application.
Collapse
Affiliation(s)
- Chenshuang Li
- Department of Orthodontics, School of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Zane Mills
- College of DentistryUniversity of OklahomaOklahoma CityOklahomaUSA
| | - Zhong Zheng
- Division of Growth and Development, School of DentistryUniversity of CaliforniaLos AngelesCaliforniaUSA
- Department of Surgery, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
10
|
Lai K, Xi Y, Du X, Jiang Z, Li Y, Huang T, Miao X, Wang H, Wang Y, Yang G. Activation of Nell-1 in BMSC Sheet Promotes Implant Osseointegration Through Regulating Runx2/Osterix Axis. Front Cell Dev Biol 2020; 8:868. [PMID: 33072736 PMCID: PMC7536315 DOI: 10.3389/fcell.2020.00868] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/11/2020] [Indexed: 12/11/2022] Open
Abstract
Neural epidermal growth factor-like 1 protein (Nell-1) is first studied because of its association with human craniosynostosis. Nell-1 has been used to accelerate the process of fracture healing because of the osteoinductive ability in recent years. However, the role of Nell-1 during the process of osteointegration is unknown. Here we show that activation of Nell-1 in the BMSC sheet promotes osseointegration in vivo and in vitro. We found that overexpression of Nell-1 improved osteogenic differentiation and enhanced matrix mineralization of BMSCs through increasing expression of Runx2 and Osterix. Activation of Nell-1 up-regulated the expression ratio of OPG/RANKL, which might have a negative influence on osteoclast differentiation. Furthermore, we obtained BMSC sheet-implant complexes transfected with lentivirus overexpressing and interfering Nell-1 in in vivo study, and confirmed that overexpression of Nell-1 promoted new bone formation around the implant and increased the bone-implant contacting area percentage. Our results demonstrate that activation of Nell-1 improves implant osteointegration by regulating Runx2/Osterix axis and shows the potential of BMSC sheet-implant complexes in gene therapy.
Collapse
Affiliation(s)
- Kaichen Lai
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Yue Xi
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Xue Du
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Zhiwei Jiang
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Yongzheng Li
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Tingben Huang
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Xiaoyan Miao
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Huiming Wang
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Ying Wang
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Guoli Yang
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| |
Collapse
|
11
|
Liu Y, Ju M, Wang Z, Li J, Shao C, Fu T, Jing Y, Zhao Y, Lv Z, Li G. The synergistic effect of NELL1 and adipose-derived stem cells on promoting bone formation in osteogenesis imperfecta treatment. Biomed Pharmacother 2020; 128:110235. [PMID: 32454289 DOI: 10.1016/j.biopha.2020.110235] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/30/2020] [Accepted: 05/03/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Osteogenesis imperfecta (OI) is a rare genetic disorder characterized by bone fragility and deformity. Mesenchymal stem cells (MSCs) infusion can improve bone performance mainly due to their differentiation into osteoblasts in OI therapy. The osteoinductive activity of NELL1 have benefited various bone defect and osteoporotic models by promoting bone formation. The present study investigated the efficacy of combined use of NELL1 and adipose-derived mesenchymal stem cells (ADSCs) in OI treatment. METHODS Lentiviral vector carrying mouse Nell1 gene was constructed and lentivirus were used to infect ADSCs. The osteogenic capacity of MC3T3-E1 and ADSCs stimulated by recombinant mouse NELL1 protein (rmNELL1) and Nell1 gene genetically modified ADSCs (lenti-Nell1-ADSCs) were estimated by real-time quantitative PCR. Thirty adult male OI type I mice with single Col1a1 gene knockout were randomly divided into five groups and received intravenously injected PBS, rmNELL1 (1.25 mg/Kg), ADSCs (2 × 105 cells per mice), rmNELL1 (1.25 mg/Kg) combined with ADSCs (2 × 105 cells per mice), or lenti-Nell1-ADSCs (2 × 105 cells per mice) respectively. Six wildtype (WT) mice served as positive control. Bone formation was examined after 4 weeks using micro-CT, histological and immunohistochemical methods. RESULTS Three osteoblast related genes of MC3T3-E1 and ADSCs were significantly up-regulated by rmNELL1 in vitro. Lenti-Nell1-ADSCs showed greatly enhanced osteogenic differentiation capacity. The infused lenti-Nell1-ADSCs could migrate to femur and differentiate into ALPL-positive cells. Systemic administration of rmNELL1 combined with ADSCs or lenti-Nell1-ADSCs markedly improved the femoral microstructure and promoted bone formation through increasing the ALPL and osteocalcin (OCN) expression, much better than mice that received single rmNELL1 or ADSCs. And Nell1 gene engineered ADSCs achieved slightly better outcomes than that of combinative use of rmNELL1 and ADSCs. CONCLUSIONS NELL1 and ADSCs exhibited synergistic effect on stimulating bone formation of OI mice, which might provide an alternative strategy in OI treatment. Compared with dose escalation or multiple administration of rmNELL1, lentivirus-mediated long term expression of NELL1 might be more feasible and convenient. However, further studies are needed to confirm the safety and optimize the therapeutic regime.
Collapse
Affiliation(s)
- Yi Liu
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Mingyan Ju
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Zihan Wang
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Jiaci Li
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Chenyi Shao
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Ting Fu
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Yaqing Jing
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Yuxia Zhao
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Zhe Lv
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Guang Li
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, People's Republic of China.
| |
Collapse
|
12
|
In situ bone tissue engineering using gene delivery nanocomplexes. Acta Biomater 2020; 108:326-336. [PMID: 32160962 DOI: 10.1016/j.actbio.2020.03.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 02/29/2020] [Accepted: 03/04/2020] [Indexed: 02/06/2023]
Abstract
Gene delivery offers promising outcomes for functional recovery or regeneration of lost tissues at cellular and tissue levels. However, more efficient carriers are needed to safely and locally delivery of genetic materials. Herein, we demonstrate microfluidic-assisted synthesis of plasmid DNA (pDNA)-based nanocomplexe (NC) platforms for bone tissue regeneration. pDNA encoding human bone morphogenesis protein-2 (BMP-2) was used as a gene of interest. Formation and fine-tuning of nanocomplexes (NCs) between pDNA and chitosan (CS) as carriers were performed using a micromixer platform. Flow characteristics were adjusted to tune mixing time and consequently size, zeta potential, and compactness of assembled NCs. Subsequently, NCs were immobilized on a nanofibrous Poly(ε-caprolactone) (PCL) scaffold functionalized with metalloprotease-sensitive peptide (MMP-sensitive). This construct can provide an environmental-sensitive and localized gene delivery platform. Osteogenic differentiation of bone marrow-derived mesenchymal stem cells (MSCs) was studied using chemical and biological assays. The presented results converge to indicate a great potential of the developed methodology for in situ bone tissue engineering using immobilized microfluidic-synthesized gene delivery nanocomplexes, which is readily expandable in the field of regenerative nanomedicine. STATEMENT OF SIGNIFICANCE: In this study, we demonstrate microfluidic-assisted synthesis of plasmid DNA (pDNA)-based nanocomplexes (NCs) platforms for bone tissue regeneration. We used pDNA encoding human bone morphogenesis protein-2 (BMP-2) as the gene of interest. Using micromixer platform nanocomplexes (NCs) between pDNA and chitosan (CS) were fabricated and optimized. NCs were immobilized on a nanofibrous polycaprolactone scaffold functionalized with metalloprotease-sensitive peptide. In vitro and in vivo assays confirmed the osteogenic differentiation of mesenchymal stem cells (MSCs). The obtained data indicated great potential of the developed methodology for in situ bone tissue engineering using immobilized microfluidic-synthesized gene delivery nanocomplexes, which is readily expandable in the field of regenerative nanomedicine.
Collapse
|
13
|
Yilmaz E, Mihci E, Nur B, Alper ÖM, Taçoy Ş. Recent Advances in Craniosynostosis. Pediatr Neurol 2019; 99:7-15. [PMID: 31421914 DOI: 10.1016/j.pediatrneurol.2019.01.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 12/25/2018] [Accepted: 01/24/2019] [Indexed: 12/27/2022]
Abstract
Craniosynostosis is a pathologic craniofacial disorder and is defined as the premature fusion of one or more cranial (calvarial) sutures. Cranial sutures are fibrous joints consisting of nonossified mesenchymal cells that play an important role in the development of healthy craniofacial skeletons. Early fusion of these sutures results in incomplete brain development that may lead to complications of several severe medical conditions including seizures, brain damage, mental delay, complex deformities, strabismus, and visual and breathing problems. As a congenital disease, craniosynostosis has a heterogeneous origin that can be affected by genetic and epigenetic alterations, teratogens, and environmental factors and make the syndrome highly complex. To date, approximately 200 syndromes have been linked to craniosynostosis. In addition to being part of a syndrome, craniosynostosis can be nonsyndromic, formed without any additional anomalies. More than 50 nuclear genes that relate to craniosynostosis have been identified. Besides genetic factors, epigenetic factors like microRNAs and mechanical forces also play important roles in suture fusion. As craniosynostosis is a multifactorial disorder, evaluating the craniosynostosis syndrome requires and depends on all the information obtained from clinical findings, genetic analysis, epigenetic or environmental factors, or gene modulators. In this review, we will focus on embryologic and genetic studies, as well as epigenetic and environmental studies. We will discuss published studies and correlate the findings with unknown aspects of craniofacial disorders.
Collapse
Affiliation(s)
- Elanur Yilmaz
- Department of Medical Biology and Genetics, Akdeniz University Medical School, Antalya, Turkey
| | - Ercan Mihci
- Department of Pediatric Genetics, Akdeniz University Medical School, Antalya, Turkey
| | - Banu Nur
- Department of Pediatric Genetics, Akdeniz University Medical School, Antalya, Turkey
| | - Özgül M Alper
- Department of Medical Biology and Genetics, Akdeniz University Medical School, Antalya, Turkey.
| | - Şükran Taçoy
- Department of Pediatric Genetics, Akdeniz University Medical School, Antalya, Turkey
| |
Collapse
|
14
|
Liu L, Lam WMR, Naidu M, Yang Z, Wang M, Ren X, Hu T, Kumarsing R, Ting K, Goh JCH, Wong HK. Synergistic Effect of NELL-1 and an Ultra-Low Dose of BMP-2 on Spinal Fusion. Tissue Eng Part A 2019; 25:1677-1689. [PMID: 31337284 DOI: 10.1089/ten.tea.2019.0124] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Bone morphogenetic protein 2 (BMP-2) is widely used in spinal fusion but it can cause adverse effects such as ectopic bone and adipose tissue in vivo. Neural epidermal growth factor like-like molecule-1 (NELL-1) has been shown to suppress BMP-2-induced adverse effects. However, no optimum carriers that control both NELL-1 and BMP-2 releases to elicit long-term bioactivity have been developed. In this study, we employed polyelectrolyte complex (PEC) as a control release carrier for NELL-1 and BMP-2. An ultra-low dose of BMP-2 synergistically functioned with NELL-1 on bone marrow mesenchymal stem cells osteogenic differentiation with greater mineralization in vitro. The osteoinductive ability of NELL-1 and an ultra-low dose of BMP-2 in PEC was investigated in rat posterolateral spinal fusion. Our results showed increased fusion rate, bone architecture, and improved bone stiffness at 8 weeks after surgery in the combination groups compared with NELL-1 or BMP-2 alone. Moreover, the formation of ectopic bone and adipose tissue was negligible in all the PEC groups. In summary, dual delivery of NELL-1 and an ultra-low dose of BMP-2 in the PEC control release carrier has greater fusion efficiency compared with BMP-2 alone and could potentially be a better alternative to the currently used BMP-2 treatments for spinal fusion. Impact Statement In this study, polyelectrolyte complex was used to absorb neural epidermal growth factor like-like molecule-1 (NELL-1) and bone morphogenetic protein 2 (BMP-2) to achieve controlled dual release. The addition of NELL-1 significantly reduced the effective dose of BMP-2 to 2.5% of its conventional dose in absorbable collagen sponge, to produce solid spinal fusion without significant adverse effects. This study was the first to identify the efficacy of combination NELL-1 and BMP-2 in a control release carrier in spinal fusion, which could be potentially used clinically to increase fusion rate and avoid the adverse effects commonly associated with conventional BMP-2.
Collapse
Affiliation(s)
- Ling Liu
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wing Moon Raymond Lam
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mathanapriya Naidu
- Cancer Science Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Zheng Yang
- NUS Tissue Engineering Program (NUSTEP), Life Science Institute, National University of Singapore, Singapore, Singapore
| | - Ming Wang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xiafei Ren
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Tao Hu
- Department of Spine Surgery, Shanghai East Hospital, Shanghai, China
| | - Ramruttun Kumarsing
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kang Ting
- Section of Orthodontics, School of Dentistry, Dental and Craniofacial Research Institute, University of California Los Angeles, Los Angeles, California
| | - James Cho-Hong Goh
- NUS Tissue Engineering Program (NUSTEP), Life Science Institute, National University of Singapore, Singapore, Singapore.,Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore
| | - Hee-Kit Wong
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Tissue Engineering Program (NUSTEP), Life Science Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|
15
|
Wu J, Wang Q, Han Q, Zhu H, Li M, Fang Y, Wang X. Effects of Nel-like molecule-1 and bone morphogenetic protein 2 combination on rat pulp repair. J Mol Histol 2019; 50:253-261. [PMID: 30937700 DOI: 10.1007/s10735-019-09822-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 03/27/2019] [Indexed: 01/09/2023]
Abstract
Nel-like molecule-1 (NELL-1) is a novel highly specific growth factor that can induce osteoblast differentiation and bone formation as well as odontoblast differentiation. Recent studies have suggested that NELL-1 can synergistically increase bone formation and regeneration with bone morphogenetic protein 2 (BMP2) and inhibit adverse effects induced by BMP2. This study aimed to evaluate the combined effects of NELL-1 and BMP2 on rat pulp repair. The experiment used healthy non-carious maxillary first molars from 60 Wistar rats. Exposed pulps were capped with NELL-1 plus BMP2, NELL-1 alone, and BMP2 alone, and each was absorbed onto a sterile collagen sponge. In the control samples, the collagen sponge alone and Dycal were used as capping agents. After l, 2 and 4 weeks, the rats were sacrificed. The formation of reparative dentin, as well the situation of pulp repair, was detected by hematoxylin-eosin (HE) staining; moreover, the expression of dentin specific protein-dentin sialophosphoprotein (DSPP) and the pro-inflammatory cytokines interleukin-6 (IL6) and interleukin-8 (IL8) was detected by immunohistochemical staining. Quantitative real-time PCR experiment was used to investigate the mRNA levels of IL6 and IL8. The results showed that pulp capping with NELL-1 plus BMP2 in rats had superior ability in inducing reparative dentin formation with dentin tubules and in reducing the inflammatory cell response compared with the other groups. These findings suggested that combined use of NELL-1 and BMP2 could positively regulate pulp repair.
Collapse
Affiliation(s)
- Jiameng Wu
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Jinan, Shandong, China
| | - Qiang Wang
- Jinan Stomatological Hospital, Jinan, 250001, Shandong, China
| | - Qi Han
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Jinan, Shandong, China
| | - Hongfan Zhu
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Jinan, Shandong, China
| | - Mengyue Li
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Jinan, Shandong, China
| | - Yixuan Fang
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Jinan, Shandong, China
| | - Xiaoying Wang
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
16
|
Qi H, Kim JK, Ha P, Chen X, Chen E, Chen Y, Li J, Pan HC, Yu M, Mohazeb Y, Azer S, Baik L, Kwak JH, Ting K, Zhang X, Hu M, Soo C. Inactivation of Nell-1 in Chondrocytes Significantly Impedes Appendicular Skeletogenesis. J Bone Miner Res 2019; 34:533-546. [PMID: 30352124 PMCID: PMC6677149 DOI: 10.1002/jbmr.3615] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 09/18/2018] [Accepted: 10/06/2018] [Indexed: 12/29/2022]
Abstract
NELL-1, an osteoinductive protein, has been shown to regulate skeletal ossification. Interestingly, an interstitial 11p14.1-p15.3 deletion involving the Nell-1 gene was recently reported in a patient with short stature and delayed fontanelle closure. Here we sought to define the role of Nell-1 in endochondral ossification by investigating Nell-1-specific inactivation in Col2α1-expressing cell lineages. Nell-1flox/flox ; Col2α1-Cre+ (Nell-1Col2α1 KO) mice were generated for comprehensive analysis. Nell-1Col2α1 KO mice were born alive but displayed subtle femoral length shortening. At 1 and 3 months postpartum, Nell-1 inactivation resulted in dwarfism and premature osteoporotic phenotypes. Specifically, Nell-1Col2α1 KO femurs and tibias exhibited significantly reduced length, bone mineral density (BMD), bone volume per tissue volume (BV/TV), trabecular number/thickness, cortical volume/thickness/density, and increased trabecular separation. The decreased bone formation rate revealed by dynamic histomorphometry was associated with altered numbers and/or function of osteoblasts and osteoclasts. Furthermore, longitudinal observations by in vivo micro-CT showed delayed and reduced mineralization at secondary ossification centers in mutants. Histologically, reduced staining intensities of Safranin O, Col-2, Col-10, and fewer BrdU-positive chondrocytes were observed in thinner Nell-1Col2α1 KO epiphyseal plates along with altered distribution and weaker expression level of Ihh, Patched-1, PTHrP, and PTHrP receptor. Primary Nell-1Col2α1 KO chondrocytes also exhibited decreased proliferation and differentiation, and its downregulated expression of the Ihh-PTHrP signaling molecules can be partially rescued by exogenous Nell-1 protein. Moreover, intranuclear Gli-1 protein and gene expression of the Gli-1 downstream target genes, Hip-1 and N-Myc, were also significantly decreased with Nell-1 inactivation. Notably, the rescue effects were diminished/reduced with application of Ihh signaling inhibitors, cyclopamine or GANT61. Taken together, these findings suggest that Nell-1 is a pivotal modulator of epiphyseal homeostasis and endochondral ossification. The cumulative chondrocyte-specific Nell-1 inactivation significantly impedes appendicular skeletogenesis resulting in dwarfism and premature osteoporosis through inhibiting Ihh signaling and predominantly altering the Ihh-PTHrP feedback loop. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Huichuan Qi
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, Changchun, Jilin, P. R. China
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Jong Kil Kim
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Pin Ha
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Xiaoyan Chen
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
- Department of Orthodontics, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Eric Chen
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Yao Chen
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Jiayi Li
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Hsin Chuan Pan
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Mengliu Yu
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
- Center of Stomatology, China-Japan Friendship Hospital, 2nd Yinghuayuan East Street, Chaoyang District, Beijing, P. R. China
| | - Yasamin Mohazeb
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Sophia Azer
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Lloyd Baik
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Jin Hee Kwak
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Kang Ting
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Xinli Zhang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Min Hu
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, Changchun, Jilin, P. R. China
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
17
|
Li C, Zheng Z, Ha P, Chen X, Jiang W, Sun S, Chen F, Asatrian G, Berthiaume EA, Kim JK, Chen EC, Pang S, Zhang X, Ting K, Soo C. Neurexin Superfamily Cell Membrane Receptor Contactin-Associated Protein Like-4 (Cntnap4) Is Involved in Neural EGFL-Like 1 (Nell-1)-Responsive Osteogenesis. J Bone Miner Res 2018; 33:1813-1825. [PMID: 29905970 PMCID: PMC6390490 DOI: 10.1002/jbmr.3524] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/29/2018] [Accepted: 06/06/2018] [Indexed: 01/28/2023]
Abstract
Contactin-associated protein-like 4 (Cntnap4) is a member of the neurexin superfamily of transmembrane molecules that have critical functions in neuronal cell communication. Cntnap4 knockout mice display decreased presynaptic gamma-aminobutyric acid (GABA) and increased dopamine release that is associated with severe, highly penetrant, repetitive, and perseverative movements commonly found in human autism spectrum disorder patients. However, no known function of Cntnap4 has been revealed besides the nervous system. Meanwhile, secretory protein neural EGFL-like 1 (Nell-1) is known to exert potent osteogenic effects in multiple small and large animal models without the off-target effects commonly found with bone morphogenetic protein 2. In this study, while searching for a Nell-1-specific cell surface receptor during osteogenesis, we identified and validated a ligand/receptor-like interaction between Nell-1 and Cntnap4 by demonstrating: 1) Nell-1 and Cntnap4 colocalization on the surface of osteogenic-committed cells; 2) high-affinity interaction between Nell-1 and Cntnap4; 3) abrogation of Nell-1-responsive Wnt and MAPK signaling transduction, as well as osteogenic effects, via Cntnap4 knockdown; and 4) replication of calvarial cleidocranial dysplasias-like defects observed in Nell-1-deficient mice in Wnt1-Cre-mediated Cntnap4-knockout transgenic mice. In aggregate, these findings indicate that Cntnap4 plays a critical role in Nell-1-responsive osteogenesis. Further, this is the first functional annotation for Cntnap4 in the musculoskeletal system. Intriguingly, Nell-1 and Cntnap4 also colocalize on the surface of human hippocampal interneurons, implicating Nell-1 as a potential novel ligand for Cntnap4 in the nervous system. This unexpected characterization of the ligand/receptor-like interaction between Nell-1 and Cntnap4 indicates a novel biological functional axis for Nell-1 and Cntnap4 in osteogenesis and, potentially, in neural development and function. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Chenshuang Li
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zhong Zheng
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Pin Ha
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xiaoyan Chen
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA.,The Affiliated Hospital of Stomatology, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Wenlu Jiang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Shan Sun
- Department of Biological Sciences and Biotechnology, State Key Laboratory of Biomembrane and Membrane Biotechnology, Tsinghua University, Beijing, PR China
| | - Feng Chen
- School and Hospital of Stomatology, Peking University, Beijing, PR China
| | - Greg Asatrian
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Emily A Berthiaume
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jong Kil Kim
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Eric C Chen
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Shen Pang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xinli Zhang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kang Ting
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA.,Division of Plastic and Reconstructive Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
18
|
Appelman-Dijkstra NM, Papapoulos SE. Clinical advantages and disadvantages of anabolic bone therapies targeting the WNT pathway. Nat Rev Endocrinol 2018; 14:605-623. [PMID: 30181608 DOI: 10.1038/s41574-018-0087-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The WNT signalling pathway is a key regulator of bone metabolism, particularly bone formation, which has helped to define the role of osteocytes - the most abundant bone cells - as orchestrators of bone remodelling. Several molecules involved in the control of the WNT signalling pathway have been identified as potential targets for the development of bone-building therapeutics for patients with osteoporosis. Several of these molecules have been investigated in animal models, but only inhibitors of sclerostin (which is produced by osteocytes) have been investigated in phase III clinical studies. Here, we review the rationale for these developments and the specificity and potential off-target actions of WNT-based therapeutics. We also describe the available preclinical and clinical studies and discuss the benefits and risks of using sclerostin inhibitors for the management of patients with osteoporosis.
Collapse
|
19
|
Zhang N, Ma L, Liu X, Jiang X, Yu Z, Zhao D, Zhang L, Zhang C, Huang F. In vitro and in vivo evaluation of xenogeneic bone putty with the carrier of hydrogel derived from demineralized bone matrix. Cell Tissue Bank 2018; 19:591-601. [PMID: 29974309 DOI: 10.1007/s10561-018-9708-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 06/29/2018] [Indexed: 02/06/2023]
Abstract
The demineralized bone matrix (DBM) putty is a traditional bone graft utilized to facilitate the repair and reconstruction of bone. Recent studies indicated the DBM putties with the various carriers were different in bone repairing ability. In order to prepare a kind of DBM putty with a good biocompatibility and bioactivity, the DBM gel was processed from the DBM and the feasibility as a carrier for the DBM putty was evaluated. After the bovine DBM gel was prepared, the BMPs content as well as the ability to promote osteogenic differentiation of MC3T3-E1 cells in vitro were investigated. Then the DBM putty was prepared and filled into the rat calvarial defect model to evaluate the bone repairing ability by micro-CT and histology. The result showed there was 2.953 ± 0.054 ng BMP contained in per gram of the DBM gel. And the ALP production of MC3T3-E1 cells in the DBM gels group increased with prolonged culturing, the mineralized nodules formed in MC3T3-E1 cells on 14th day after co-culture. The putty prepared by DBM gel was easy to handle without loss of DBM particles at room temperature. In the rat calvarial bone defect experiment, histological observation showed more mature bone formed in the DBM putty group than that in the type I collagen group at 12 weeks, which indicated the bone putty prepared by DBM gel exhibited a better bone repair capability.
Collapse
Affiliation(s)
- Naili Zhang
- Department of Human Anatomy, School of Basic Medical Science, Binzhou Medical University, 346 Guanhai Road, Laishan, Yantai, 264003, Shandong, China
| | - Lina Ma
- Department of Diagnostics, School of Medicine, Binzhou Medical University, 346 Guanhai Road, Laishan, Yantai, 264003, Shandong, China
| | - Xiaowei Liu
- Department of Human Anatomy, School of Basic Medical Science, Binzhou Medical University, 346 Guanhai Road, Laishan, Yantai, 264003, Shandong, China
| | - Xiaorui Jiang
- Department of Hand and Foot Surgery, Yuhuangding Hospital, 20 Yuhuangding East Road, Zhifu, Yantai, 264000, China
| | - Zhenhai Yu
- Department of Human Anatomy, School of Basic Medical Science, Binzhou Medical University, 346 Guanhai Road, Laishan, Yantai, 264003, Shandong, China
| | - Dongmei Zhao
- Department of Human Anatomy, School of Basic Medical Science, Binzhou Medical University, 346 Guanhai Road, Laishan, Yantai, 264003, Shandong, China
| | - Luping Zhang
- Department of Human Anatomy, School of Basic Medical Science, Binzhou Medical University, 346 Guanhai Road, Laishan, Yantai, 264003, Shandong, China
| | - Chunlei Zhang
- Department of Human Anatomy, School of Basic Medical Science, Binzhou Medical University, 346 Guanhai Road, Laishan, Yantai, 264003, Shandong, China
| | - Fei Huang
- Department of Human Anatomy, School of Basic Medical Science, Binzhou Medical University, 346 Guanhai Road, Laishan, Yantai, 264003, Shandong, China.
| |
Collapse
|
20
|
Chen H, Zhang Z, Zhang L, Wang J, Zhang M, Zhu B. miR-27a protects human mitral valve interstitial cell from TNF-α-induced inflammatory injury via up-regulation of NELL-1. ACTA ACUST UNITED AC 2018; 51:e6997. [PMID: 29694513 PMCID: PMC5937725 DOI: 10.1590/1414-431x20186997] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 01/18/2018] [Indexed: 02/06/2023]
Abstract
MicroRNAs (miRNAs) have been reported to be associated with heart valve disease, which can be caused by inflammation. This study aimed to investigate the functional impacts of miR-27a on TNF-α-induced inflammatory injury in human mitral valve interstitial cells (hMVICs). hMVICs were subjected to 40 ng/mL TNF-α for 48 h, before which the expressions of miR-27a and NELL-1 in hMVICs were altered by stable transfection. Trypan blue staining, BrdU incorporation assay, flow cytometry detection, ELISA, and western blot assay were performed to detect cell proliferation, apoptosis, and the release of proinflammatory cytokines. We found that miR-27a was lowly expressed in response to TNF-α exposure in hMVICs. Overexpression of miR-27a rescued hMVICs from TNF-α-induced inflammatory injury, as cell viability and BrdU incorporation were increased, apoptotic cell rate was decreased, Bcl-2 was up-regulated, Bax and cleaved caspase-3/9 were down-regulated, and the release of IL-1β, IL-6, and MMP-9 were reduced. NELL-1 was positively regulated by miR-27a, and NELL-1 up-regulation exhibited protective functions during TNF-α-induced cell damage. Furthermore, miR-27a blocked JNK and Wnt/β-catenin signaling pathways, and the blockage was abolished when NELL-1 was silenced. This study demonstrated that miR-27a overexpression protected hMVICs from TNF-α-induced cell damage, which might be via up-regulation of NELL-1 and thus modulation of JNK and Wnt/β-catenin signaling pathways.
Collapse
Affiliation(s)
- Honglei Chen
- Department of Cardiology, Chengyang People's Hospital, Qingdao, Shandong, China
| | - Zhixu Zhang
- Department of Cardiology, Chengyang People's Hospital, Qingdao, Shandong, China
| | - Li Zhang
- Teaching and Research Office of Immunology, Qingdao University, Qingdao, Shandong, China
| | - Junzhi Wang
- Department of Cardiology, Chengyang People's Hospital, Qingdao, Shandong, China
| | - Minghui Zhang
- Department of Imaging, Eastern District of Linyi People's Hospital, Linyi, Shandong, China
| | - Bin Zhu
- Department of Cardiology, Chengyang People's Hospital, Qingdao, Shandong, China
| |
Collapse
|
21
|
Zhao H, Qin X, Zhang Q, Zhang X, Lin J, Ting K, Chen F. Nell-1-ΔE, a novel transcript of Nell-1, inhibits cell migration by interacting with enolase-1. J Cell Biochem 2018; 119:5725-5733. [PMID: 29388706 DOI: 10.1002/jcb.26756] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 01/25/2018] [Indexed: 11/06/2022]
Abstract
NELL-1 is a secreted protein that was originally found to be upregulated in pathologically fusing and fused sutures in non-syndromic unilateral coronal synostosis patients. Apart from the ability of NELL-1 to promote osteogenesis in long and craniofacial bones, NELL-1 reportedly inhibits the formation of several benign and malignant tumors. We previously identified a novel transcript of Nell-1 that lacked a calcium-binding epidermal growth factor (EGF)-like domain compared with full-length Nell-1; this new transcript was named Nell-1-ΔE. Three obvious structural differences between these two isoforms were revealed by homology modeling. Furthermore, the recombinant Nell-1-ΔE protein, but not the full-length Nell-1 protein, inhibited cell migration in vitro. However, full-length Nell-1 and Nell-1-ΔE proteins were present in similar subcellular locations and displayed similar expression patterns in both the intracellular and extracellular spaces. The results from the co-immunoprecipitation and liquid chromatography/tandem mass spectrometry analyses using two cell lines demonstrated that Nell-1-ΔE but not full-length Nell-1 interacted with enolase-1 in the extracellular spaces of both cell lines. The results of wound healing assays using ENO-1-overexpressing cells treated with full-length Nell-1/Nell-1-ΔE suggested that Nell-1-ΔE inhibited cell migration by interacting with ENO-1. Our study indicated that the novel transcript Nell-1-ΔE, but not full-length Nell-1, might be a candidate tumor suppressor factor for basic research and clinical practice.
Collapse
Affiliation(s)
- Huaxiang Zhao
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, P. R. China
| | - Xueyan Qin
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, P. R. China
| | - Qian Zhang
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, P. R. China
| | - Xinli Zhang
- Dental and Craniofacial Research Institute, University of California, Los Angeles, California
| | - Jiuxiang Lin
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, P. R. China
| | - Kang Ting
- Dental and Craniofacial Research Institute, University of California, Los Angeles, California
| | - Feng Chen
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, P. R. China
| |
Collapse
|
22
|
Tanjaya J, Lord EL, Wang C, Zhang Y, Kim JK, Nguyen A, Baik L, Pan HC, Chen E, Kwak JH, Zhang X, Wu B, Soo C, Ting K. The Effects of Systemic Therapy of PEGylated NEL-Like Protein 1 (NELL-1) on Fracture Healing in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:715-727. [PMID: 29294300 PMCID: PMC5840496 DOI: 10.1016/j.ajpath.2017.11.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 11/04/2017] [Accepted: 11/21/2017] [Indexed: 01/05/2023]
Abstract
Fractures are common, with an incidence of 13.7 per 1000 adults annually. Systemic agents have been widely used for enhancing bone regeneration; however, the efficacy of these therapeutics for the management and prevention of fracture remains unclear. NEL-like protein 1 (NELL-1) is a potent pro-osteogenic cytokine that has been modified with polyethylene glycol (PEG)ylation [PEGylated NELL-1 (NELL-PEG)] to enhance its pharmacokinetics for systemic therapy. Our aim was to investigate the effects of systemic administration of NELL-PEG on fracture healing in mice and on overall bone properties in uninjured bones. Ten-week-old CD-1 mice were subjected to an open osteotomy of bilateral radii and treated with weekly injections of NELL-PEG or PEG phosphate-buffered saline as control. Systemic injection of NELL-PEG resulted in improved bone mineral density of the fracture site and accelerated callus union. After 4 weeks of treatment, mice treated with NELL-PEG exhibited substantially enhanced callus volume, callus mineralization, and biomechanical properties. NELL-PEG injection significantly augmented bone regeneration, as confirmed by high expression of bone turnover rate, bone formation rate, and mineral apposition rate. Consistently, the immunohistochemistry results also confirmed a high bone remodeling activity in the NELL-PEG-treated group. Our findings suggest that weekly injection of NELL-PEG may have the clinical potential to accelerate fracture union and enhance overall bone properties, which may help prevent subsequent fractures.
Collapse
Affiliation(s)
- Justine Tanjaya
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Elizabeth L Lord
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Chenchao Wang
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; Department of Plastic Surgery, First Hospital of China Medical University, Shenyang, China
| | - Yulong Zhang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California; Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, California
| | - Jong K Kim
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Alan Nguyen
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Llyod Baik
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Hsin C Pan
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Eric Chen
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Jin H Kwak
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Xinli Zhang
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Benjamin Wu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California; Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, California
| | - Chia Soo
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California; Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Kang Ting
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
23
|
Chitosan-stablized bovine serum albumin nanoparticles having ability to control the release of NELL-1 protein. Int J Biol Macromol 2017; 109:672-680. [PMID: 29288032 DOI: 10.1016/j.ijbiomac.2017.12.104] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/19/2017] [Accepted: 12/19/2017] [Indexed: 11/24/2022]
Abstract
The study was designed to prepare and evaluate chitosan stabilized-albumin nanoparticles as NELL-1 protein carriers(Chi/NNPs). The Chi/NNPs were prepared by desolvation method and then stabilized by chitosan through electrostatic interaction. The Chi/NNPs were characterized for drug loading efficiency, surface morphology, particle size, surface charge. Fluorescein isothiocyanate-labeled chitosan was used to confirm the homogeneity of chitosan coating on the BSA nanoparticles. The NELL-1 bioactivity of Chi/NNPs and the release kinetics were investigated in vitro. It was observed that the mean particle size with chitosan (0.075 wt%,0.15 wt%, 0.3 wt%, respectively) and the surface charge were 368.663 ± 15.470 nm, 382.881 ± 18.767 nm, 390.480 ± 11.465 nm and +25.03 ± 1.42 mV, +30.27 ± 1.80 mV, +31.03 ± 2.05 mV respectively. Drug entrapment efficiency ranged from 87.83% to 89.30%. The Chi/NNPs prepared with the 0.15 wt% chitosan were able to successfully control the release of NELL-1 and maintain a sustained release for up to 8 days. Furthermore, more than 82.67 ± 8.74% of the loaded protein's bioactivity was preserved in Chi/NNPs over the period of the investigation. Our findings suggest that Chi/NNPs as promising protein delivery nanocarriers have the ability to maintain sustained release kinetics and to preserve the bioactivity of released NELL-1.
Collapse
|
24
|
Fahmy-Garcia S, van Driel M, Witte-Buoma J, Walles H, van Leeuwen JPTM, van Osch GJVM, Farrell E. NELL-1, HMGB1, and CCN2 Enhance Migration and Vasculogenesis, But Not Osteogenic Differentiation Compared to BMP2. Tissue Eng Part A 2017; 24:207-218. [PMID: 28463604 DOI: 10.1089/ten.tea.2016.0537] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Currently, autografts still represent the gold standard treatment for the repair of large bone defects. However, these are associated with donor-site morbidity and increased pain, cost, and recovery time. The ideal therapy would use biomaterials combined with bone growth factors to induce and instruct bone defect repair without the need to harvest patient tissue. In this line, bone morphogenetic proteins (BMPs) have been the most extensively used agents for clinical bone repair, but at supraphysiological doses that are not without risk. Because of the need to eliminate the risks of BMP2 use in vivo, we assessed the ability of three putative osteogenic factors, nel-like molecule type 1 (NELL-1), high mobility group box 1 (HMGB1), and CCN2, to enhance the essential processes for bone defect repair in vitro and compared them to BMP2. Although it has been reported that NELL-1, HMGB1, and CCN2 play a role in bone formation, less is known about the contribution of these proteins to the different events involved, such as cell migration, osteogenesis, and vasculogenesis. In this study, we investigated the effects of different doses of NELL-1, HMGB, CCN2, and BMP2 on these three processes as a model for the recruitment and differentiation of resident cells in the in vivo bone defect repair situation, using cells of human origin. Our data demonstrated that NELL-1, HMGB1, and CCN2 significantly induced mesenchymal stem cell migration (from 1.58-fold increase compared to control), but BMP2 did not. Interestingly, only BMP2 increased osteogenesis in marrow stromal cells, whereas it inhibited osteogenesis in preosteoblasts. Moreover, the four proteins studied promoted significantly endothelial cell migration, reaching a maximum of 2.4-fold increase compared to control, and induced formation of tube-like structures. NELL-1, HMGB1, and CCN2 had these effects at relatively low doses compared to BMP2. This work indicates that NELL-1, HMGB1, and CCN2 might enhance bone defect healing via the recruitment of endogenous cells and induction of vascularization and act via different processes than BMP2.
Collapse
Affiliation(s)
| | | | - Janneke Witte-Buoma
- 3 Department of Oral and Maxillofacial Surgery, Erasmus MC , Rotterdam, The Netherlands
| | - Heike Walles
- 4 Department Tissue Engineering and Regenerative Medicine, University Hospital Würzburg , Würzburg, Germany
| | | | - Gerjo J V M van Osch
- 1 Department of Orthopaedics, Erasmus MC , Rotterdam, The Netherlands .,5 Otorhinolaryngology Department, Erasmus MC, Rotterdam, The Netherlands
| | - Eric Farrell
- 3 Department of Oral and Maxillofacial Surgery, Erasmus MC , Rotterdam, The Netherlands
| |
Collapse
|
25
|
Pakvasa M, Alverdy A, Mostafa S, Wang E, Fu L, Li A, Oliveira L, Athiviraham A, Lee MJ, Wolf JM, He TC, Ameer GA, Reid RR. Neural EGF-like protein 1 (NELL-1): Signaling crosstalk in mesenchymal stem cells and applications in regenerative medicine. Genes Dis 2017; 4:127-137. [PMID: 29276737 PMCID: PMC5737940 DOI: 10.1016/j.gendis.2017.07.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 07/27/2017] [Indexed: 12/15/2022] Open
Abstract
Bone tissue regeneration holds the potential to solve both osteoporosis and large skeletal defects, two problems associated with significant morbidity. The differentiation of mesenchymal stem cells into the osteogenic lineage requires a specific microenvironment and certain osteogenic growth factors. Neural EGF Like-Like molecule 1 (NELL-1) is a secreted glycoprotein that has proven, both in vitro and in vivo, to be a potent osteo-inductive factor. Furthermore, it has been shown to repress adipogenic differentiation and inflammation. NELL-1 can work synergistically with other osteogenic factors such as Bone Morphogenic Protein (BMP) -2 and -9, and has shown promise for use in tissue engineering and as a systemically administered drug for the treatment of osteoporosis. Here we provide a comprehensive up-to-date review on the molecular signaling cascade of NELL-1 in mesenchymal stem cells and potential applications in bone regenerative engineering.
Collapse
Affiliation(s)
- Mikhail Pakvasa
- The University of Chicago, Pritzker School of Medicine, Chicago, IL 60637, USA
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Alex Alverdy
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Rosalind Franklin University, Chicago Medical School, North Chicago, IL 60064, USA
| | - Sami Mostafa
- The University of Chicago, Pritzker School of Medicine, Chicago, IL 60637, USA
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Eric Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Lucy Fu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Alexander Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Leonardo Oliveira
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Aravind Athiviraham
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael J. Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jennifer Moriatis Wolf
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Guillermo A. Ameer
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Russell R. Reid
- The University of Chicago, Pritzker School of Medicine, Chicago, IL 60637, USA
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| |
Collapse
|
26
|
Abstract
BACKGROUND Nonhealing bone defects represent an immense biomedical burden. Despite recent advances in protein-based bone regeneration, safety concerns over bone morphogenetic protein-2 have prompted the search for alternative factors. Previously, the authors examined the additive/synergistic effects of hedgehog and Nel-like protein-1 (NELL-1) on the osteogenic differentiation of mesenchymal stem cells in vitro. In this study, the authors sought to leverage their previous findings by applying the combination of Smoothened agonist (SAG), hedgehog signal activator, and NELL-1 to an in vivo critical-size bone defect model. METHODS A 4-mm parietal bone defect was created in mixed-gender CD-1 mice. Treatment groups included control (n = 6), SAG (n = 7), NELL-1 (n = 7), and SAG plus NELL-1 (n = 7). A custom fabricated poly(lactic-co-glycolic acid) disk with hydroxyapatite coating was used as an osteoinductive scaffold. RESULTS Results at 4 and 8 weeks showed increased bone formation by micro-computed tomographic analyses with either stimulus alone (SAG or NELL-1), but significantly greater bone formation with both components combined (SAG plus NELL-1). This included greater bone healing scores and increased bone volume and bone thickness. Histologic analyses confirmed a significant increase in new bone formation with the combination therapy SAG plus NELL-1, accompanied by increased defect vascularization. CONCLUSIONS In summary, the authors' results suggest that combining the hedgehog signaling agonist SAG and NELL-1 has potential as a novel therapeutic strategy for the healing of critical-size bone defects. Future directions will include optimization of dosage and delivery strategy for an SAG and NELL-1 combination product.
Collapse
|
27
|
Ren Z, Ma S, Jin L, Liu Z, Liu D, Zhang X, Cai Q, Yang X. Repairing a bone defect with a three-dimensional cellular construct composed of a multi-layered cell sheet on electrospun mesh. Biofabrication 2017. [PMID: 28631613 DOI: 10.1088/1758-5090/aa747f] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
In addition to providing maneuverability, electrospun nanofibrous meshes can make excellent supports for constructing flexible cell sheets to regulate cell behavior by nanofiber features. With the target of bone regeneration, herein composite nanofibers with two different fiber arrangements (nestlike, random) were electrospun from a blend solution containing poly(l-lactide) (PLLA) and gelatin (1:1 in weight ratio). Unlike the non-woven morphology in a random nanofibrous mesh, PLLA/gelatin composite nanofibers in the nestlike nanofibrous mesh displayed both non-woven and parallel morphologies. Both kinds of nanofibrous mesh were ∼50 μm thick as-prepared, and shrank to ∼30 μm after seeding with bone mesenchymal stromal cells (BMSCs). After 7 days of in vitro culture, cell sheets could form on both meshes (CSM) and on the culture plate. It was found that application of nanofibrous mesh promoted the osteogenic differentiation of BMSC sheets compared with the control. The nestlike mesh displayed slight superiority over the random mesh in enhancing osteogenic differentiation, but their different fiber arrangements did not cause much difference in cell proliferation. Three-dimensional multi-layered CSM constructs were built by stacking four mono-layered CSMs together. The CSM constructs (based on a nestlike or random nanofibrous mesh) were incubated in vitro for 3 days before being implanted into rat cranial defects. In comparison with the control group, there was significant formation of new calcified bone in both CSM construct-filled groups at 12 weeks' post-operation. The nestlike group showed slightly better bone healing (based on both qualitative and quantitative analysis) than the random group, while showing insignificant differences. We showed that the concept of using a three-dimensional multi-layered CSM construct in enhancing bone regeneration was feasible. Future studies should take more nanofiber features (e.g. bioactive components) into account to further enhance osteogenesis.
Collapse
Affiliation(s)
- Zhiwei Ren
- State Key Laboratory of Organic-Inorganic Composites; Beijing Laboratory of Biomedical Materials; Beijing University of Chemical Technology, Beijing 100029, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
28
|
James AW, Shen J, Tsuei R, Nguyen A, Khadarian K, Meyers CA, Pan HC, Li W, Kwak JH, Asatrian G, Culiat CT, Lee M, Ting K, Zhang X, Soo C. NELL-1 induces Sca-1+ mesenchymal progenitor cell expansion in models of bone maintenance and repair. JCI Insight 2017; 2:92573. [PMID: 28614787 PMCID: PMC5470886 DOI: 10.1172/jci.insight.92573] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 05/05/2017] [Indexed: 12/25/2022] Open
Abstract
NELL-1 is a secreted, osteogenic protein first discovered to control ossification of the cranial skeleton. Recently, NELL-1 has been implicated in bone maintenance. However, the cellular determinants of NELL-1's bone-forming effects are still unknown. Here, recombinant human NELL-1 (rhNELL-1) implantation was examined in a clinically relevant nonhuman primate lumbar spinal fusion model. Prolonged rhNELL-1 protein release was achieved using an apatite-coated β-tricalcium phosphate carrier, resulting in a local influx of stem cell antigen-1-positive (Sca-1+) mesenchymal progenitor cells (MPCs), and complete osseous fusion across all samples (100% spinal fusion rate). Murine studies revealed that Nell-1 haploinsufficiency results in marked reductions in the numbers of Sca-1+CD45-CD31- bone marrow MPCs associated with low bone mass. Conversely, rhNELL-1 systemic administration in mice showed a marked anabolic effect accompanied by increased numbers of Sca-1+CD45-CD31- bone marrow MPCs. Mechanistically, rhNELL-1 induces Sca-1 transcription among MPCs, in a process requiring intact Wnt/β-catenin signaling. In summary, NELL-1 effectively induces bone formation across small and large animal models either via local implantation or intravenous delivery. NELL-1 induces an expansion of a bone marrow subset of MPCs with Sca-1 expression. These findings provide compelling justification for the clinical translation of a NELL-1-based therapy for local or systemic bone formation.
Collapse
Affiliation(s)
- Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA.,UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California, USA
| | - Jia Shen
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, USA
| | - Rebecca Tsuei
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, USA
| | - Alan Nguyen
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, USA
| | - Kevork Khadarian
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, USA
| | - Carolyn A Meyers
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hsin Chuan Pan
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, USA
| | - Weiming Li
- Department of Orthopedics, The First Clinical Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jin H Kwak
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, USA
| | - Greg Asatrian
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, USA
| | | | - Min Lee
- Section of Biomaterials, School of Dentistry, UCLA, Los Angeles, California, USA
| | - Kang Ting
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, USA
| | - Xinli Zhang
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, USA
| | - Chia Soo
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, California, USA.,Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| |
Collapse
|
29
|
Zhang Y, Dong R, Park Y, Bohner M, Zhang X, Ting K, Soo C, Wu BM. Controlled release of NELL-1 protein from chitosan/hydroxyapatite-modified TCP particles. Int J Pharm 2016; 511:79-89. [PMID: 27349789 PMCID: PMC6705139 DOI: 10.1016/j.ijpharm.2016.06.050] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 06/15/2016] [Accepted: 06/19/2016] [Indexed: 01/18/2023]
Abstract
NEL-like molecule-1 (NELL-1) is a novel osteogenic protein that showing high specificity to osteochondral cells. It was widely used in bone regeneration research by loading onto carriers such as tricalcium phosphate (TCP) particles. However, there has been little research on protein controlled release from this material and its potential application. In this study, TCP was first modified with a hydroxyapatite coating followed by a chitosan coating to prepare chitosan/hydroxyapatite-coated TCP particles (Chi/HA-TCP). The preparation was characterized by SEM, EDX, FTIR, XRD, FM and Zeta potential measurements. The NELL-1 loaded Chi/HA-TCP particles and the release kinetics were investigated in vitro. It was observed that the Chi/HA-TCP particles prepared with the 0.3% (wt/wt) chitosan solution were able to successfully control the release of NELL-1 and maintain a slow, steady release for up to 28 days. Furthermore, more than 78% of the loaded protein's bioactivity was preserved in Chi/HA-TCP particles over the period of the investigation, which was significantly higher than that of the protein released from hydroxyapatite coated TCP (HA-TCP) particles. Collectively, this study suggests that the osteogenic protein NELL-1 showed a sustained release pattern after being encapsulated into the modified Chi/HA-TCP particles, and the NELL-1 integrated composite of Chi/HA-TCP showed a potential to function as a protein delivery carrier and as an improved bone matrix for use in bone regeneration research.
Collapse
Affiliation(s)
- Yulong Zhang
- Department of Bioengineering, Department of Materials Science and Engineering, and Division of Advanced Prosthodontics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rui Dong
- Beijing Institute of Dental Research, School of Stomatology, Capital Medical University, Beijing, 100050, PR China
| | - Yujin Park
- Department of Bioengineering, Department of Materials Science and Engineering, and Division of Advanced Prosthodontics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Marc Bohner
- RMS Foundation, Bischmattstr. 12, CH-2544 Bettlach, Switzerland
| | - Xinli Zhang
- Weintraub Center for Reconstructive Biotechnology, and Dental and Craniofacial Research Institute, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kang Ting
- Weintraub Center for Reconstructive Biotechnology, and Dental and Craniofacial Research Institute, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chia Soo
- Weintraub Center for Reconstructive Biotechnology, and Dental and Craniofacial Research Institute, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Benjamin M Wu
- Department of Bioengineering, Department of Materials Science and Engineering, and Division of Advanced Prosthodontics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Weintraub Center for Reconstructive Biotechnology, and Dental and Craniofacial Research Institute, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
30
|
Tanjaya J, Zhang Y, Lee S, Shi J, Chen E, Ang P, Zhang X, Tetradis S, Ting K, Wu B, Soo C, Kwak JH. Efficacy of Intraperitoneal Administration of PEGylated NELL-1 for Bone Formation. Biores Open Access 2016; 5:159-70. [PMID: 27354930 PMCID: PMC4921932 DOI: 10.1089/biores.2016.0018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Systemically delivered NEL-like molecule-1 (NELL-1), a potent pro-osteogenic protein, promotes bone formation in healthy and osteoporotic mouse models. PEGylation of NELL-1 (NELL-PEG) increases the half-life of the protein in a mouse model without compromising its osteogenic potential, thereby improving its pharmacokinetics upon systemic delivery. This study consists of a twofold approach: a biodistribution test and an in vivo osteogenic potential test. The biodistribution test compared two commonly used administration methods for drug delivery other than intravenous-intraperitoneal (IP) and subcutaneous (SC)-to examine NELL-PEG biodistribution in mice. Compared to a single-dose SC injection (1.25 mg/kg), a single-dose IP administration yielded a higher protein uptake in the targeted bone sites. When the IP injection dose was doubled to 2.5 mg/kg, the protein remained in the femurs, tibias, and vertebrae for up to 72 h. Next, based on the results of the biodistribution study, IP administration was selected to further investigate the in vivo osteogenic effects of weekly NELL-PEG injection (q7d). In vivo, the IP administered NELL-PEG group showed significantly greater bone mineral density, bone volume fraction, and trabecular bone formation in the targeted bone sites compared to the phosphate-buffered saline control. In summary, weekly NELL-PEG injection via IP administration successfully enhanced the overall bone quality. These findings demonstrate that systemic delivery of NELL-PEG via IP administration may serve as an effective osteogenic therapy for preventing and treating osteoporosis.
Collapse
Affiliation(s)
- Justine Tanjaya
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Yulong Zhang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California.; Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, California
| | - Soonchul Lee
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University, Pocheon, South Korea.; Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Jiayu Shi
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Eric Chen
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Pia Ang
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California.; Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Xinli Zhang
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Sotirios Tetradis
- Section of Oral and Maxillofacial Radiology, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Kang Ting
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California.; Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Benjamin Wu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California.; Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, California
| | - Chia Soo
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California.; Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Jin Hee Kwak
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| |
Collapse
|
31
|
James AW, Chiang M, Asatrian G, Shen J, Goyal R, Chung CG, Chang L, Shrestha S, Turner AS, Seim HB, Zhang X, Wu BM, Ting K, Soo C. Vertebral Implantation of NELL-1 Enhances Bone Formation in an Osteoporotic Sheep Model. Tissue Eng Part A 2016; 22:840-9. [PMID: 27113550 DOI: 10.1089/ten.tea.2015.0230] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Vertebral compression fractures related to osteoporosis greatly afflict the aging population. One of the most commonly used therapy today is balloon kyphoplasty. However, this treatment is far from ideal and is associated with significant side effects. NELL-1, an osteoinductive factor that possesses both pro-osteogenic and anti-osteoclastic properties, is a promising candidate for an alternative to current treatment modalities. This study utilizes the pro-osteogenic properties of recombinant human NELL-1 (rhNELL-1) in lumbar spine vertebral defect model in osteoporotic sheep. METHODS Osteoporosis was induced through ovariectomy, dietary depletion of calcium and vitamin D, and steroid administration. After osteoporotic induction, lumbar vertebral body defect creation was performed. Sheep were randomly implanted with the control vehicle, comprised of hyaluronic acid (HA) with hydroxyapatite-coated β-tricalcium phosphate (β-TCP), or the treatment material of rhNELL-1 protein lyophilized onto β-TCP mixed with HA. Analysis of lumbar spine defect healing was performed by radiographic, histologic, and computer-simulated biomechanical testing. RESULTS rhNELL-1 treatment significantly increased lumbar spine bone formation, as determined by bone mineral density, % bone volume, and mean cortical width as assessed by micro-computed tomography. Histological analysis revealed a significant increase in bone area and osteoblast number and decrease in osteoclast number around the implant site. Computer-simulated biomechanical analysis of trabecular bone demonstrated that rhNELL-1-treatment resulted in a significantly more stress-resistant composition. CONCLUSION Our findings suggest rhNELL-1-based vertebral implantation successfully improved cortical and cancellous bone regeneration in the lumbar spine of osteoporotic sheep. rhNELL-1-based bone graft substitutes represent a potential new local therapy.
Collapse
Affiliation(s)
- Aaron W James
- 1 Departments of Surgery and Orthopaedic Surgery, Orthopaedic Hospital Research Center, UCLA and Orthopedic Hospital , Los Angeles, California.,2 Division of Growth and Development and Section of Orthodontics, Dental and Craniofacial Research Institute, School of Dentistry, University of California , Los Angeles, Los Angeles, California.,3 Department of Pathology and Laboratory Medicine, University of California , Los Angeles, Los Angeles, California
| | - Michael Chiang
- 2 Division of Growth and Development and Section of Orthodontics, Dental and Craniofacial Research Institute, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Greg Asatrian
- 2 Division of Growth and Development and Section of Orthodontics, Dental and Craniofacial Research Institute, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Jia Shen
- 1 Departments of Surgery and Orthopaedic Surgery, Orthopaedic Hospital Research Center, UCLA and Orthopedic Hospital , Los Angeles, California.,2 Division of Growth and Development and Section of Orthodontics, Dental and Craniofacial Research Institute, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Raghav Goyal
- 2 Division of Growth and Development and Section of Orthodontics, Dental and Craniofacial Research Institute, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Choon G Chung
- 2 Division of Growth and Development and Section of Orthodontics, Dental and Craniofacial Research Institute, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Le Chang
- 1 Departments of Surgery and Orthopaedic Surgery, Orthopaedic Hospital Research Center, UCLA and Orthopedic Hospital , Los Angeles, California.,2 Division of Growth and Development and Section of Orthodontics, Dental and Craniofacial Research Institute, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Swati Shrestha
- 1 Departments of Surgery and Orthopaedic Surgery, Orthopaedic Hospital Research Center, UCLA and Orthopedic Hospital , Los Angeles, California.,2 Division of Growth and Development and Section of Orthodontics, Dental and Craniofacial Research Institute, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - A Simon Turner
- 4 Department of Veterinary Sciences, Colorado State University , Fort Collins, Colorado
| | - Howard B Seim
- 4 Department of Veterinary Sciences, Colorado State University , Fort Collins, Colorado
| | - Xinli Zhang
- 1 Departments of Surgery and Orthopaedic Surgery, Orthopaedic Hospital Research Center, UCLA and Orthopedic Hospital , Los Angeles, California.,2 Division of Growth and Development and Section of Orthodontics, Dental and Craniofacial Research Institute, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Benjamin M Wu
- 5 Departments of Bioengineering and Material Sciences, University of California , Los Angeles, Los Angeles, California
| | - Kang Ting
- 1 Departments of Surgery and Orthopaedic Surgery, Orthopaedic Hospital Research Center, UCLA and Orthopedic Hospital , Los Angeles, California.,2 Division of Growth and Development and Section of Orthodontics, Dental and Craniofacial Research Institute, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Chia Soo
- 1 Departments of Surgery and Orthopaedic Surgery, Orthopaedic Hospital Research Center, UCLA and Orthopedic Hospital , Los Angeles, California.,6 Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California , Los Angeles, Los Angeles, California
| |
Collapse
|
32
|
Jang JW, Lee JS, Jung UW, Kim CS, Cho KS. In Vivo Evaluation of Commercially Available Gel-Type Polyethylene Glycol Membrane for Carrier of Recombinant Human Bone Morphogenetic Protein-2. J Oral Maxillofac Surg 2016; 75:297.e1-297.e13. [PMID: 27288839 DOI: 10.1016/j.joms.2016.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 05/10/2016] [Accepted: 05/11/2016] [Indexed: 11/15/2022]
Abstract
PURPOSE This study evaluated a commercially available, 3-dimensional gel-type polyethylene glycol (PEG) membrane as a carrier for recombinant human bone morphogenetic protein-2 (rhBMP-2) using a rat calvarial defect model. Another gel-type carrier, fibrin-fibronectin system (FFS), was used as a positive control. MATERIALS AND METHODS Critical-size defects were made in the rat calvarium, which were allocated to 1 of 10 groups comprising 2 healing periods and biomaterial conditions: 1) sham control, 2) FFS only, 3) FFS plus BMP-2, 4) PEG only, and 5) PEG plus BMP-2. Radiographic and histologic analyses were performed at 2 and 8 weeks after surgery. RESULTS After 2 weeks, some parts of the FFS were biodegraded and extensive cellular infiltration was observed at sites that received FFS or FFS plus BMP-2. The PEG membrane retained its augmented volume without cellular infiltration at sites that received PEG or PEG plus BMP-2. After 8 weeks, the FFS was completely degraded and replaced by new bone and connective tissues. In contrast, the volume of residual PEG was similar to that at 2 weeks, with slight cellular infiltration. In particular, there was progressive bone regeneration around micro-cracks and resorbed outer surface in the PEG + BMP-2 group. Although the PEG + BMP-2 group showed increased area and percentage of new bone, there was no statistical relevance after 2 and 8 weeks in histomorphometric analyses. However, the appearance of the healing differed (with new bone formation along micro-cracks in the PEG + BMP-2 group), and further studies with longer healing periods are needed to draw conclusions about clinical applications. CONCLUSION Evidence of mechanical stability and new bone formation along micro-cracks when using PEG plus BMP-2 might support the PEG membrane as a candidate carrier material for rhBMP-2.
Collapse
Affiliation(s)
- Ji-Woong Jang
- PhD Student, Department of Periodontology, Research Institute for Periodontal Regeneration, College of Dentistry, Yonsei University, Seoul, Korea
| | - Jung-Seok Lee
- Clinical Assistant Professor, Department of Periodontology, Research Institute for Periodontal Regeneration, College of Dentistry, Yonsei University, Seoul, Korea
| | - Ui-Won Jung
- Associate Professor, Department of Periodontology, Research Institute for Periodontal Regeneration, College of Dentistry, Yonsei University, Seoul, Korea
| | - Chang-Sung Kim
- Professor and Department Head, Department of Periodontology, Research Institute for Periodontal Regeneration, College of Dentistry, Yonsei University, Seoul, Korea
| | - Kyoo-Sung Cho
- Professor, Department of Periodontology, Research Institute for Periodontal Regeneration, College of Dentistry, Yonsei University, Seoul, Korea.
| |
Collapse
|
33
|
Pang S, Shen J, Liu Y, Chen F, Zheng Z, James AW, Hsu CY, Zhang H, Lee KS, Wang C, Li C, Chen X, Jia H, Zhang X, Soo C, Ting K. Proliferation and osteogenic differentiation of mesenchymal stem cells induced by a short isoform of NELL-1. Stem Cells 2015; 33:904-15. [PMID: 25376942 DOI: 10.1002/stem.1884] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 09/19/2014] [Accepted: 09/27/2014] [Indexed: 01/11/2023]
Abstract
Neural epidermal growth factor-like (NEL)-like protein 1 (NELL-1) has been identified as an osteoinductive differentiation factor that promotes mesenchymal stem cell (MSC) osteogenic differentiation. In addition to full-length NELL-1, there are several NELL-1-related transcripts reported. We used rapid amplification of cDNA ends to recover potential cDNA of NELL-1 isoforms. A NELL-1 isoform with the N-terminal 240 amino acid (aa) residues truncated was identified. While full-length NELL-1 that contains 810 aa residues (NELL-1810 ) plays an important role in embryologic skeletal development, the N-terminal-truncated NELL-1 isoform (NELL-1570 ) was expressed postnatally. Similar to NELL-1810 , NELL-1570 induced MSC osteogenic differentiation. In addition, NELL-1570 significantly stimulated MSC proliferation in multiple MSC-like populations such as murine C3H10T1/2 MSC cell line, mouse primary MSCs, and perivascular stem cells, which is a type of stem cells proposed as the perivascular origin of MSCs. In contrast, NELL-1810 demonstrated only limited stimulation of MSC proliferation. Similar to NELL-1810 , NELL-1570 was found to be secreted from host cells. Both NELL-1570 expression lentiviral vector and column-purified recombinant protein NELL-1570 demonstrated almost identical effects in MSC proliferation and osteogenic differentiation, suggesting that NELL-1570 may function as a pro-osteogenic growth factor. In vivo, NELL-1570 induced significant calvarial defect regeneration accompanied by increased cell proliferation. Thus, NELL-1570 has the potential to be used for cell-based or hormone-based therapy of bone regeneration.
Collapse
Affiliation(s)
- Shen Pang
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Shen J, LaChaud G, Shrestha S, Asatrian G, Zhang X, Dry SM, Soo C, Ting K, James AW. NELL-1 expression in tumors of cartilage. J Orthop 2015; 12:S223-9. [PMID: 27047227 DOI: 10.1016/j.jor.2015.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 10/04/2015] [Indexed: 10/22/2022] Open
Abstract
BACKGROUND/AIMS NELL-1 is a novel osteochondral differentiation factor protein with increasing usage in tissue engineering. Previously, we reported the expression patterns of NELL-1 in bone-forming skeletal tumors. With increasing interest in the use of NELL-1 protein, we sought to examine the expression of NELL-1 in cartilage-forming tumors. METHODS Immunohistochemical expression was examined in human pathologic specimens. RESULTS Consistent NELL-1 overexpression across all cartilage-forming tumors was observed. Similar degrees of expression were observed in enchondroma, chondrosarcoma, and chondroblastic osteosarcoma. NELL-1 expression did not significantly vary by tumor grade. CONCLUSION In summary, NELL-1 demonstrates reliable and consistent expression across cartilage-forming skeletal tumors.
Collapse
Affiliation(s)
- Jia Shen
- Division of Growth and Development and Section of Orthodontics, School of Dentistry UCLA, Los Angeles, CA 90095, United States; Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA 90095, United States
| | - Gregory LaChaud
- Division of Growth and Development and Section of Orthodontics, School of Dentistry UCLA, Los Angeles, CA 90095, United States; Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA 90095, United States; Vanderbilt University School of Medicine, Nashville, TN 37212, United States
| | - Swati Shrestha
- Division of Growth and Development and Section of Orthodontics, School of Dentistry UCLA, Los Angeles, CA 90095, United States; Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA 90095, United States
| | - Greg Asatrian
- Division of Growth and Development and Section of Orthodontics, School of Dentistry UCLA, Los Angeles, CA 90095, United States; Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA 90095, United States
| | - Xinli Zhang
- Division of Growth and Development and Section of Orthodontics, School of Dentistry UCLA, Los Angeles, CA 90095, United States
| | - Sarah M Dry
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA 90095, United States
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, Department of Surgery, UCLA, Los Angeles, CA 90095, United States; UCLA and Orthopaedic Hospital, Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, CA 90095, United States
| | - Kang Ting
- Division of Growth and Development and Section of Orthodontics, School of Dentistry UCLA, Los Angeles, CA 90095, United States
| | - Aaron W James
- Division of Growth and Development and Section of Orthodontics, School of Dentistry UCLA, Los Angeles, CA 90095, United States; Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA 90095, United States; UCLA and Orthopaedic Hospital, Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, Los Angeles, CA 90095, United States; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, United States
| |
Collapse
|
35
|
James AW, Shen J, Zhang X, Asatrian G, Goyal R, Kwak JH, Jiang L, Bengs B, Culiat CT, Turner AS, Seim Iii HB, Wu BM, Lyons K, Adams JS, Ting K, Soo C. NELL-1 in the treatment of osteoporotic bone loss. Nat Commun 2015; 6:7362. [PMID: 26082355 PMCID: PMC4557288 DOI: 10.1038/ncomms8362] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 04/28/2015] [Indexed: 01/09/2023] Open
Abstract
NELL-1 is a secreted, osteoinductive protein whose expression rheostatically controls skeletal ossification. Overexpression of NELL-1 results in craniosynostosis in humans and mice, whereas lack of Nell-1 expression is associated with skeletal undermineralization. Here we show that Nell-1-haploinsufficient mice have normal skeletal development but undergo age-related osteoporosis, characterized by a reduction in osteoblast:osteoclast (OB:OC) ratio and increased bone fragility. Recombinant NELL-1 binds to integrin β1 and consequently induces Wnt/β-catenin signalling, associated with increased OB differentiation and inhibition of OC-directed bone resorption. Systemic delivery of NELL-1 to mice with gonadectomy-induced osteoporosis results in improved bone mineral density. When extended to a large animal model, local delivery of NELL-1 to osteoporotic sheep spine leads to significant increase in bone formation. Altogether, these findings suggest that NELL-1 deficiency plays a role in osteoporosis and demonstrate the potential utility of NELL-1 as a combination anabolic/antiosteoclastic therapeutic for bone loss.
Collapse
Affiliation(s)
- Aaron W James
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA and Orthopaedic Hospital, University of California, Los Angeles, California 90095, USA.,Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California 90095, USA.,Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA
| | - Jia Shen
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA and Orthopaedic Hospital, University of California, Los Angeles, California 90095, USA.,Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California 90095, USA
| | - Xinli Zhang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California 90095, USA
| | - Greg Asatrian
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California 90095, USA
| | - Raghav Goyal
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California 90095, USA
| | - Jin H Kwak
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California 90095, USA
| | - Lin Jiang
- Department of Neurology, Easton Center for Alzheimer's Disease Research, Molecular Biology Institute, University of California, Los Angeles, California 90095, USA
| | - Benjamin Bengs
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA and Orthopaedic Hospital, University of California, Los Angeles, California 90095, USA
| | | | - A Simon Turner
- Department of Veterinary Sciences, Colorado State University, Fort Collins, Colorado 80523, USA
| | - Howard B Seim Iii
- Department of Veterinary Sciences, Colorado State University, Fort Collins, Colorado 80523, USA
| | - Benjamin M Wu
- Department of Bioengineering and Department of Material Sciences, University of California, Los Angeles, California 90095, USA
| | - Karen Lyons
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA and Orthopaedic Hospital, University of California, Los Angeles, California 90095, USA
| | - John S Adams
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA and Orthopaedic Hospital, University of California, Los Angeles, California 90095, USA
| | - Kang Ting
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA and Orthopaedic Hospital, University of California, Los Angeles, California 90095, USA.,Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California 90095, USA
| | - Chia Soo
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA and Orthopaedic Hospital, University of California, Los Angeles, California 90095, USA.,Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
36
|
Abstract
Normal bone healing is a complex process that eventually restores original structure and function to the site of trauma. However, clinical circumstances such as nonunion, critical-sized defects, systemic bone disease, and fusion procedures have stimulated a search for ways to enhance this normal healing process. Biologics are an important part of this search and many, including bone marrow aspirate concentrate, demineralized bone matrix, platelet-rich plasma, bone morphogenic proteins, and platelet-derived growth factor, are currently in clinical use. Many others, including mesenchymal stem cells, parathyroid hormone, and Nel-like molecule-1 (NELL-1) will likely be in use in the future depending on the results of preclinical and clinical trials.
Collapse
Affiliation(s)
- Benjamin Smith
- Department of Orthopedic Surgery and Orthopedic Research Laboratory, Feinstein Institute for Medical Research and North Shore-LIJ Health System, Manhasset, NY, USA,
| | | | | |
Collapse
|
37
|
Fan J, Im CS, Cui ZK, Guo M, Bezouglaia O, Fartash A, Lee JY, Nguyen J, Wu BM, Aghaloo T, Lee M. Delivery of Phenamil Enhances BMP-2-Induced Osteogenic Differentiation of Adipose-Derived Stem Cells and Bone Formation in Calvarial Defects. Tissue Eng Part A 2015; 21:2053-65. [PMID: 25869476 DOI: 10.1089/ten.tea.2014.0489] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) have been widely used for bone repair in the craniofacial region. However, its high dose requirement in clinical applications revealed adverse effects and inefficient bone formation, along with high cost. Here, we report a novel osteoinductive strategy to effectively complement the osteogenic activity of BMP-2 using phenamil, a small molecule that can induce osteogenic differentiation via stimulation of BMP signaling. Treatment of adipose-derived stem cells (ASCs) with BMP-2 in combination with phenamil significantly promoted the in vitro osteogenic differentiation of ASCs. The efficacy of the combination strategy of phenamil+BMP-2 was further confirmed in a mouse calvarial defect model using scaffolds consisting of poly(lactic-co-glycolic acid) and apatite layer on their surfaces designed to slowly release phenamil and BMP-2. Six weeks after implantation, the scaffolds treated with phenamil+BMP-2 significantly promoted mouse calvarial regeneration as demonstrated by micro-computerized tomography and histology, compared with the groups treated with phenamil or BMP-2 alone. Moreover, the combination treatment reduced the BMP-2 dose without compromising calvarial healing efficacy. These results suggest promising complementary therapeutic strategies for bone repair in more efficient and cost-effective manners.
Collapse
Affiliation(s)
- Jiabing Fan
- 1 Division of Advanced Prosthodontics, UCLA School of Dentistry , Los Angeles, California
| | - Choong Sung Im
- 1 Division of Advanced Prosthodontics, UCLA School of Dentistry , Los Angeles, California
| | - Zhong-Kai Cui
- 1 Division of Advanced Prosthodontics, UCLA School of Dentistry , Los Angeles, California
| | - Mian Guo
- 2 Department of Neurosurgery, the 2nd Affiliated Hospital of Harbin Medical University , Harbin, China
| | - Olga Bezouglaia
- 3 Division of Diagnostic and Surgical Sciences, UCLA School of Dentistry , Los Angeles, California
| | - Armita Fartash
- 3 Division of Diagnostic and Surgical Sciences, UCLA School of Dentistry , Los Angeles, California
| | - Ju-Yeon Lee
- 1 Division of Advanced Prosthodontics, UCLA School of Dentistry , Los Angeles, California
| | - John Nguyen
- 1 Division of Advanced Prosthodontics, UCLA School of Dentistry , Los Angeles, California
| | - Benjamin M Wu
- 4 Department of Bioengineering, University of California , Los Angeles, California
| | - Tara Aghaloo
- 3 Division of Diagnostic and Surgical Sciences, UCLA School of Dentistry , Los Angeles, California
| | - Min Lee
- 1 Division of Advanced Prosthodontics, UCLA School of Dentistry , Los Angeles, California.,4 Department of Bioengineering, University of California , Los Angeles, California
| |
Collapse
|
38
|
Pharmacokinetics and osteogenic potential of PEGylated NELL-1 in vivo after systemic administration. Biomaterials 2015; 57:73-83. [PMID: 25913252 DOI: 10.1016/j.biomaterials.2015.03.063] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 03/28/2015] [Accepted: 03/31/2015] [Indexed: 12/11/2022]
Abstract
Osteoporosis is a skeletal disorder attributable to an imbalance in osteoblast and osteoclast activity. NELL-1, a secretory protein that promotes osteogenesis while suppressing osteoclastic activity, holds potential as an osteoporosis therapy. Recently, we demonstrated that PEGylation of NELL-1 significantly improves its thermostability while preserving its bioactivity in vitro. However, the effect of PEGylation on the pharmacokinetics and osteogenic potential of NELL-1 in vivo have yet to be investigated. The present study demonstrated that PEGylation of NELL-1 significantly increases the elimination half-life time of the protein from 5.5 h to 15.5 h while distributing more than 2-3 times the amount of protein to bone tissues (femur, tibia, vertebrae, calvaria) in vivo when compared to naked NELL-1. In addition, microCT and DXA analyses demonstrated that systemic NELL-PEG therapy administered every 4 or 7 days significantly increases not only femoral and lumbar BMD and percent bone volume, but also new bone formation throughout the overall skeleton after four weeks of treatment. Furthermore, immunohistochemistry revealed increased osteocalcin expression, while TRAP staining showed reduced osteoclast numbers in NELL-PEG groups. Our findings suggest that the PEGylation technique presents a viable and promising approach to further develop NELL-1 into an effective systemic therapeutic for the treatment of osteoporosis.
Collapse
|
39
|
Hong JY, Kim MS, Lim HC, Lee JS, Choi SH, Jung UW. A high concentration of recombinant human bone morphogenetic protein-2 induces low-efficacy bone regeneration in sinus augmentation: a histomorphometric analysis in rabbits. Clin Oral Implants Res 2015; 27:e199-e205. [PMID: 25899917 DOI: 10.1111/clr.12603] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2015] [Indexed: 12/01/2022]
Abstract
OBJECTIVES The aim of the study was to elucidate the efficacy of bone regeneration at the early stage of healing in rabbit sinuses grafted with a biphasic calcium phosphate (BCP) carrier soaked in a high concentration of recombinant human bone morphogenetic protein-2 (rhBMP-2). MATERIALS AND METHODS Both maxillary sinuses of eight male rabbits were used. The sinus on one side (assigned randomly) was grafted with BCP loaded with rhBMP-2 (1.5 mg/ml; test group) using a soaking method, while the other was grafted with saline-soaked BCP (control group). After a 2-week healing period, the sinuses were analyzed by micro-computed tomography and histomorphometry. RESULTS The total augmented area and soft tissue space were significantly larger in the test group than in the control group, whereas the opposite was true for the area of residual material and newly formed bone. Most of the new bone in the test group was localized to the Schneiderian membrane (SM), while very little bone formation was observed in the window and center regions of the sinus. New bone was distributed evenly in the control group sinuses. CONCLUSION Within the limitations of this study, it appeared that application of a high concentration of rhBMP-2 soaked onto a BCP carrier inhibited bone regeneration from the pristine bone and increased soft tissue swelling and inflammatory response at the early healing stage of sinus augmentation, although osteoinductive potential was found along the SM.
Collapse
Affiliation(s)
- Ji-Youn Hong
- Department of Periodontology, School of Dentistry, Kyung Hee University, Seoul, Korea
| | - Min-Soo Kim
- Department of Periodontology, Research Institute for Periodontal Regeneration, College of Dentistry, Yonsei University, Seoul, South Korea
| | - Hyun-Chang Lim
- Department of Periodontology, School of Dentistry, Kyung Hee University, Seoul, Korea
| | - Jung-Seok Lee
- Department of Periodontology, Research Institute for Periodontal Regeneration, College of Dentistry, Yonsei University, Seoul, South Korea
| | - Seong-Ho Choi
- Department of Periodontology, Research Institute for Periodontal Regeneration, College of Dentistry, Yonsei University, Seoul, South Korea
| | - Ui-Won Jung
- Department of Periodontology, Research Institute for Periodontal Regeneration, College of Dentistry, Yonsei University, Seoul, South Korea
| |
Collapse
|
40
|
Nakamura R, Oyama T, Tajiri R, Mizokami A, Namiki M, Nakamoto M, Ooi A. Expression and regulatory effects on cancer cell behavior of NELL1 and NELL2 in human renal cell carcinoma. Cancer Sci 2015; 106:656-64. [PMID: 25726761 PMCID: PMC4452169 DOI: 10.1111/cas.12649] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 02/07/2015] [Accepted: 02/25/2015] [Indexed: 12/27/2022] Open
Abstract
Neural epidermal growth factor-like like (NELL) 1 and 2 constitute a family of multimeric and multimodular extracellular glycoproteins. Although the osteogenic effects of NELL1 and functions of NELL2 in neural development have been reported, their expression and functions in cancer are largely unknown. In this study, we examined expression of NELL1 and NELL2 in renal cell carcinoma (RCC) using clinical specimens and cell lines. We show that, whereas NELL1 and NELL2 proteins are strongly expressed in renal tubules in non-cancerous areas of RCC specimens, their expression is significantly downregulated in cancerous areas. Silencing of NELL1 and NELL2 mRNA expression was also detected in RCC cell lines. Analysis of NELL1/2 promoter methylation status indicated that the CpG islands in the NELL1 and NELL2 genes are hypermethylated in RCC cell lines. NELL1 and NELL2 bind to RCC cells, suggesting that these cells express a receptor for NELL1 and NELL2 that can transduce signals. Furthermore, we found that both NELL1 and NELL2 inhibit RCC cell migration, and NELL1 further inhibits RCC cell adhesion. These results suggest that silencing of NELL gene expression by promoter hypermethylation plays roles in RCC progression by affecting cancer cell behavior.
Collapse
Affiliation(s)
- Ritsuko Nakamura
- Department of Molecular and Cellular Pathology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Takeru Oyama
- Department of Molecular and Cellular Pathology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Ryosuke Tajiri
- Department of Molecular and Cellular Pathology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Atsushi Mizokami
- Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Mikio Namiki
- Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Masaru Nakamoto
- Aberdeen Developmental Biology Group, School of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Akishi Ooi
- Department of Molecular and Cellular Pathology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| |
Collapse
|
41
|
Shen J, LaChaud G, Khadarian K, Shrestha S, Zhang X, Soo C, Ting K, Dry SM, James AW. NELL-1 expression in benign and malignant bone tumors. Biochem Biophys Res Commun 2015; 460:368-74. [PMID: 25791475 DOI: 10.1016/j.bbrc.2015.03.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Accepted: 03/08/2015] [Indexed: 12/31/2022]
Abstract
NELL-1 (NEL-like Protein 1) is an osteoinductive protein with increasing usage as a bone graft substitute in preclinical animal models. NELL-1 was first identified to have bone-forming properties by its overexpression in fusing cranial sutures. Since this time, addition of recombinant NELL-1 has been used to successfully induce bone formation in the calvarial, axial and appendicular skeleton. With increasing interest in the use of NELL-1 as a bone-graft substitute, we sought to examine the expression of NELL-1 in a wide spectrum of benign and malignant bone-forming skeletal tumors. Immunohistochemical expression was examined in human pathologic specimens. Quantitative RT-PCR evaluated NELL-1 expression among OS cell lines in vitro. Results showed NELL-1 expression in all bone tumors. Likewise, all OS cell lines demonstrated increased NELL-1 expression in comparison to non-lesional human bone marrow stromal cells. Among, benign bone tumors (osteoid osteoma and osteoblastoma), strong and diffuse staining was observed, which spatially correlated with markers of osteogenic differentiation. In contrast, a relative reduction in NELL-1 staining was observed in osteosarcoma, accompanied by increased variation between tumors. Among osteosarcoma specimens, NELL-1 expression did not correlate well with markers of osteogenic differentiation. Surprisingly, among osteosarcoma subtypes, fibroblastic osteosarcoma demonstrated the highest expression of NELL-1. In summary, NELL-1 demonstrates diffuse and reliable expression in benign but not malignant bone-forming skeletal tumors. Future studies will further define the basic biologic, diagnostic and prognostic importance of NELL-1 in bone neoplasms.
Collapse
Affiliation(s)
- Jia Shen
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California, David Geffen School of Medicine, 10833 Le Conte Ave., 13-145 CHS, Los Angeles, CA 90095, USA; Department of Pathology and Laboratory Medicine, University of California, David Geffen School of Medicine, 10833 Le Conte Ave., 13-145 CHS, Los Angeles, CA 90095, USA
| | - Greg LaChaud
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California, David Geffen School of Medicine, 10833 Le Conte Ave., 13-145 CHS, Los Angeles, CA 90095, USA; Department of Pathology and Laboratory Medicine, University of California, David Geffen School of Medicine, 10833 Le Conte Ave., 13-145 CHS, Los Angeles, CA 90095, USA
| | - Kevork Khadarian
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California, David Geffen School of Medicine, 10833 Le Conte Ave., 13-145 CHS, Los Angeles, CA 90095, USA
| | - Swati Shrestha
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California, David Geffen School of Medicine, 10833 Le Conte Ave., 13-145 CHS, Los Angeles, CA 90095, USA; Department of Pathology and Laboratory Medicine, University of California, David Geffen School of Medicine, 10833 Le Conte Ave., 13-145 CHS, Los Angeles, CA 90095, USA
| | - Xinli Zhang
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California, David Geffen School of Medicine, 10833 Le Conte Ave., 13-145 CHS, Los Angeles, CA 90095, USA
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, David Geffen School of Medicine, 10833 Le Conte Ave., 13-145 CHS, Los Angeles, CA 90095, USA; UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and The Orthopaedic Hospital Research Center, University of California, David Geffen School of Medicine, 10833 Le Conte Ave., 13-145 CHS, Los Angeles, CA 90095, USA
| | - Kang Ting
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California, David Geffen School of Medicine, 10833 Le Conte Ave., 13-145 CHS, Los Angeles, CA 90095, USA
| | - Sarah M Dry
- Department of Pathology and Laboratory Medicine, University of California, David Geffen School of Medicine, 10833 Le Conte Ave., 13-145 CHS, Los Angeles, CA 90095, USA
| | - Aaron W James
- Division of Growth and Development and Section of Orthodontics, School of Dentistry, University of California, David Geffen School of Medicine, 10833 Le Conte Ave., 13-145 CHS, Los Angeles, CA 90095, USA; Department of Pathology and Laboratory Medicine, University of California, David Geffen School of Medicine, 10833 Le Conte Ave., 13-145 CHS, Los Angeles, CA 90095, USA; UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and The Orthopaedic Hospital Research Center, University of California, David Geffen School of Medicine, 10833 Le Conte Ave., 13-145 CHS, Los Angeles, CA 90095, USA.
| |
Collapse
|
42
|
Kyllönen L, D’Este M, Alini M, Eglin D. Local drug delivery for enhancing fracture healing in osteoporotic bone. Acta Biomater 2015; 11:412-34. [PMID: 25218339 DOI: 10.1016/j.actbio.2014.09.006] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 08/30/2014] [Accepted: 09/04/2014] [Indexed: 01/08/2023]
Abstract
Fragility fractures can cause significant morbidity and mortality in patients with osteoporosis and inflict a considerable medical and socioeconomic burden. Moreover, treatment of an osteoporotic fracture is challenging due to the decreased strength of the surrounding bone and suboptimal healing capacity, predisposing both to fixation failure and non-union. Whereas a systemic osteoporosis treatment acts slowly, local release of osteogenic agents in osteoporotic fracture would act rapidly to increase bone strength and quality, as well as to reduce the bone healing period and prevent development of a problematic non-union. The identification of agents with potential to stimulate bone formation and improve implant fixation strength in osteoporotic bone has raised hope for the fast augmentation of osteoporotic fractures. Stimulation of bone formation by local delivery of growth factors is an approach already in clinical use for the treatment of non-unions, and could be utilized for osteoporotic fractures as well. Small molecules have also gained ground as stable and inexpensive compounds to enhance bone formation and tackle osteoporosis. The aim of this paper is to present the state of the art on local drug delivery in osteoporotic fractures. Advantages, disadvantages and underlying molecular mechanisms of different active species for local bone healing in osteoporotic bone are discussed. This review also identifies promising new candidate molecules and innovative approaches for the local drug delivery in osteoporotic bone.
Collapse
|
43
|
Liu J, Chen W, Zhao Z, Xu HH. Effect of NELL1 gene overexpression in iPSC-MSCs seeded on calcium phosphate cement. Acta Biomater 2014; 10:5128-5138. [PMID: 25220281 DOI: 10.1016/j.actbio.2014.08.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 08/05/2014] [Accepted: 08/15/2014] [Indexed: 02/08/2023]
Abstract
Human induced pluripotent stem cell-derived mesenchymal stem cells (iPSC-MSCs) are a promising source of patient-specific stem cells with great regenerative potential. There has been no report on NEL-like protein 1 (NELL1) gene modification of iPSC-MSCs. The objectives of this study were to genetically modify iPSC-MSCs with NELL1 overexpression for bone tissue engineering, and investigate the osteogenic differentiation of NELL1 gene-modified iPSC-MSCs seeded on Arg-Gly-Asp (RGD)-grafted calcium phosphate cement (CPC) scaffold. Cells were transduced with red fluorescence protein (RFP-iPSC-MSCs) or NELL1 (NELL1-iPSC-MSCs) by a lentiviral vector. Cell proliferation on RGD-grafted CPC scaffold, osteogenic differentiation and bone mineral synthesis were evaluated. RFP-iPSC-MSCs stably expressed high levels of RFP. Both the NELL1 gene and NELL1 protein levels were confirmed higher in NELL1-iPSC-MSCs than in RFP-iPSC-MSCs using RT-PCR and Western blot (P<0.05). Alkaline phosphatase activity was increased by 130% by NELL1 overexpression at 14days (P<0.05), indicating that NELL1 promoted iPSC-MSC osteogenic differentiation. When seeded on RGD-grafted CPC, NELL1-iPSC-MSCs attached and expanded similarly well to RFP-iPSC-MSCs. At 14days, the runt-related transcription factor 2 (RUNX2) gene level of NELL1-iPSC-MSCs was 2.0-fold that of RFP-iPSC-MSCs. The osteocalcin (OC) level of NELL1-iPSC-MSCs was 3.1-fold that of RFP-iPSC-MSCs (P<0.05). The collagen type I alpha 1 (COL1A1) gene level of NELL1-iPSC-MSCs was 1.7-fold that of RFP-iPSC-MSCs at 7days (P<0.05). Mineral synthesis was increased by 81% in NELL1-iPSC-MSCs at 21days. In conclusion, NELL1 overexpression greatly enhanced the osteogenic differentiation and mineral synthesis of iPSC-MSCs on RGD-grafted CPC scaffold for the first time. The novel NELL1-iPSC-MSC seeded RGD-CPC construct is promising for enhancing bone engineering.
Collapse
|
44
|
A Longitudinal Low Dose μCT Analysis of Bone Healing in Mice: A Pilot Study. Adv Orthop 2014; 2014:791539. [PMID: 25431676 PMCID: PMC4241339 DOI: 10.1155/2014/791539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 10/13/2014] [Indexed: 02/07/2023] Open
Abstract
Low dose microcomputed tomography (μCT) is a recently matured technique that enables the study of longitudinal bone healing and the testing of experimental treatments for bone repair. This imaging technique has been used for studying craniofacial repair in mice but not in an orthopedic context. This is mainly due to the size of the defects (approximately 1.0 mm) in long bone, which heal rapidly and may thus negatively impact the assessment of the effectiveness of experimental treatments. We developed a longitudinal low dose μCT scan analysis method combined with a new image segmentation and extraction software using Hounsfield unit (HU) scores to quantitatively monitor bone healing in small femoral cortical defects in live mice. We were able to reproducibly quantify bone healing longitudinally over time with three observers. We used high speed intramedullary reaming to prolong healing in order to circumvent the rapid healing typical of small defects. Bone healing prolongation combined with μCT imaging to study small bone defects in live mice thus shows potential as a promising tool for future preclinical research on bone healing.
Collapse
|
45
|
Abstract
Osteopenia and osteoporosis are common manifestations in inflammatory bowel diseases (IBD) but the pathogenetic mechanism of bone loss in IBD is only partially understood. There is evidence that fat mass is an important determinant of the bone mineral density and adipose-derived factors seem to play an important role for the association between fat mass and bone mass. The association between adiposity and low bone density is rather poorly studied in IBD, but emerging data on adipokines in IBD in relation to osteoporosis provide a novel pathophysiological concept that may shed light on the etiology of bone loss in IBD. It could be suggested that adipokines interfere in bone metabolism by altering the sensitive balance between osteoblasts and osteoclasts although further studies in this setting are needed.
Collapse
|
46
|
Zhang Y, Velasco O, Zhang X, Ting K, Soo C, Wu BM. Bioactivity and circulation time of PEGylated NELL-1 in mice and the potential for osteoporosis therapy. Biomaterials 2014; 35:6614-21. [PMID: 24818884 PMCID: PMC4077898 DOI: 10.1016/j.biomaterials.2014.04.061] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 04/16/2014] [Indexed: 12/12/2022]
Abstract
Osteoporosis is a progressive bone disease due to low osteoblast activity and/or high osteoclast activity. NELL-1 is a potential therapy for osteoporosis because it specifically increases osteoblast differentiation. However, similar to other protein drugs, the bioavailability of NELL-1 may be limited by its in vivo half-life and rapid clearance from body. The purpose of the present study is to prolong NELL-1 circulation time in vivo by PEGylation with three monomeric PEG sizes (5, 20, 40 kDa). While linear PEG 5k yielded the most efficient PEGylation and the most thermally stable conjugate, linear PEG 20k resulted in the conjugate with the highest Mw and longest in vivo circulation. Compared to non-modified NELL-1, all three PEGylated conjugates showed enhanced thermal stability and each prolonged the in vivo circulation time significantly. Furthermore, PEGylated NELL-1 retained its osteoblastic activity without any appreciable cytotoxicity. These findings motivate further studies to evaluate the efficacy of PEGylated NELL-1 on the prevention and treatment of osteoporosis.
Collapse
Affiliation(s)
- Yulong Zhang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Materials Science and Engineering, and Division of Advanced Prosthodontics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Omar Velasco
- Weintraub Center for Reconstructive Biotechnology, and Dental and Craniofacial Research Institute, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xinli Zhang
- Weintraub Center for Reconstructive Biotechnology, and Dental and Craniofacial Research Institute, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kang Ting
- Weintraub Center for Reconstructive Biotechnology, and Dental and Craniofacial Research Institute, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chia Soo
- Weintraub Center for Reconstructive Biotechnology, and Dental and Craniofacial Research Institute, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Benjamin M Wu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Materials Science and Engineering, and Division of Advanced Prosthodontics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Weintraub Center for Reconstructive Biotechnology, and Dental and Craniofacial Research Institute, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
47
|
Fan J, Park H, Lee MK, Bezouglaia O, Fartash A, Kim J, Aghaloo T, Lee M. Adipose-derived stem cells and BMP-2 delivery in chitosan-based 3D constructs to enhance bone regeneration in a rat mandibular defect model. Tissue Eng Part A 2014; 20:2169-79. [PMID: 24524819 PMCID: PMC4137352 DOI: 10.1089/ten.tea.2013.0523] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 02/10/2014] [Indexed: 12/29/2022] Open
Abstract
Reconstructing segmental mandiblular defects remains a challenge in the clinic. Tissue engineering strategies provide an alternative option to resolve this problem. The objective of the present study was to determine the effects of adipose-derived stem cells (ASCs) and bone morphogenetic proteins-2 (BMP-2) in three-dimensional (3D) scaffolds on mandibular repair in a small animal model. Noggin expression levels in ASCs were downregulated by a lentiviral short hairpin RNA strategy to enhance ASC osteogenesis (ASCs(Nog-)). Chitosan (CH) and chondroitin sulfate (CS), natural polysaccharides, were fabricated into 3D porous scaffolds, which were further modified with apatite coatings for enhanced cellular responses and efficient delivery of BMP-2. The efficacy of 3D apatite-coated CH/CS scaffolds supplemented with ASCs(Nog-) and BMP-2 were evaluated in a rat critical-sized mandibular defect model. After 8 weeks postimplantation, the scaffolds treated with ASCs(Nog-) and BMP-2 significantly promoted rat mandibular regeneration as demonstrated by micro-computerized tomography, histology, and immunohistochemistry, compared with the groups treated with ASCs(Nog-) or BMP-2 alone. These results suggest that our combinatorial strategy of ASCs(Nog-)+BMP-2 in 3D apatite microenvironments can significantly promote mandibular regeneration, and these may provide a potential tissue engineering approach to repair large bony defects.
Collapse
Affiliation(s)
- Jiabing Fan
- Division of Advanced Prosthodontics, UCLA School of Dentistry, Los Angeles, California
| | - Hyejin Park
- Division of Advanced Prosthodontics, UCLA School of Dentistry, Los Angeles, California
| | - Matthew K. Lee
- Department of Head and Neck Surgery, University of California, Los Angeles, Los Angeles, California
| | - Olga Bezouglaia
- Division of Diagnostic and Surgical Sciences, UCLA School of Dentistry, Los Angeles, California
| | - Armita Fartash
- Division of Diagnostic and Surgical Sciences, UCLA School of Dentistry, Los Angeles, California
| | - Jinku Kim
- Department of Bio and Chemical Engineering, Hongik University, Sejong, Korea
| | - Tara Aghaloo
- Division of Diagnostic and Surgical Sciences, UCLA School of Dentistry, Los Angeles, California
| | - Min Lee
- Division of Advanced Prosthodontics, UCLA School of Dentistry, Los Angeles, California
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
48
|
Lee JS, Lee SK, Kim BS, Im GI, Cho KS, Kim CS. Controlled release of BMP-2 using a heparin-conjugated carrier system reduces in vivo adipose tissue formation. J Biomed Mater Res A 2014; 103:545-54. [PMID: 24764177 DOI: 10.1002/jbm.a.35207] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 04/12/2014] [Accepted: 04/22/2014] [Indexed: 11/10/2022]
Abstract
There is growing concern about unwanted effects associated with the clinical use of recombinant human bone morphogenetic protein-2 (rhBMP-2) at high concentrations, including cyst-like bone formation and excessive fatty marrow formation. We, therefore, evaluated the induction of mineralized/adipose tissue formation and the bone-healing pattern associated with the controlled release of E. coli-derived rhBMP-2 (ErhBMP-2) by a heparin-conjugated fibrin (HCF) system using ectopic and orthotopic in vivo models, respectively. In the ectopic transplantation model, mineralized tissue formed at the most superficial layer of the transplanted area and on the surfaces of grafted materials, and most of the interstitial space within the transplanted area was filled with excessive adipose tissue specifically at sites that received ErhBMP-2. However, sites that received ErhBMP-2 and HCF showed significantly increased mineralized tissue formation and decreased adipose tissue formation compared to the normal fibrin system with ErhBMP-2. In the orthotopic (calvarial defect) model, controlled release of ErhBMP-2 induced by HCF significantly reduced adipose tissue formation within the defect area compared to the clinically approved absorbable collagen sponge. From these results, it can be concluded that the use of a HCF system loaded with ErhBMP-2 may reduce adipose tissue formation and enhance mineralized tissue formation.
Collapse
Affiliation(s)
- Jung-Seok Lee
- Department of Periodontology, Research Institute for Periodontal Regeneration, College of Dentistry, Yonsei University, Seoul, 120-752, Republic of Korea
| | | | | | | | | | | |
Collapse
|
49
|
Li M, Li CZ. High glucose improves healing of periodontal wound by inhibiting proliferation and osteogenetic differentiation of human PDL cells. Int Wound J 2014; 13:39-43. [PMID: 24581427 DOI: 10.1111/iwj.12218] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 12/16/2013] [Accepted: 12/19/2013] [Indexed: 01/09/2023] Open
Abstract
Periodontal ligament (PDL) cells play an important role in wound healing of periodontal tissues. Response of PDL cells' cellular activity to high-glucose concentration levels may be the key in understanding the relationship between periodontal disease and diabetes mellitus. We studied the effect of high-glucose medium on proliferation of PDL cells in vitro. PDL cells were cultured for 1, 4, 7, 10, 14 and 17 days in normal (1100 mg/l) glucose or in high (4500 mg/l) glucose medium. The 3-(4,5-dimethylithiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay for proliferation was performed. In order to evaluate the osteogenetic differentiation of human PDL cells, the cells were induced with normal- or high-glucose medium for 1, 7, 14, 21 and 28 days. The results indicated that high glucose significantly inhibited proliferation of PDL cells. Concerning the mineralised nodule formation, the percentage of calcified area to total culture dish of PDL cells in high glucose level was lower than that in normal glucose medium. The increase in alkaline phosphatase activity and collagen expression could be observed in high-glucose-containing osteogenetic factor. In conclusion, high glucose improves healing of periodontal wound by inhibiting proliferation and differentiation of PDL cells, which could explain for delayed periodontal regeneration and healing in diabetic patients.
Collapse
Affiliation(s)
- Min Li
- School & Hospital of Stomatology, Wuhan University, Hubei, Wuhan, 430079, China.,The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Hubei, Wuhan, 430079, China
| | - Cheng-Zhang Li
- School & Hospital of Stomatology, Wuhan University, Hubei, Wuhan, 430079, China.,The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Hubei, Wuhan, 430079, China
| |
Collapse
|
50
|
Nakamura Y, Hasebe A, Takahashi K, Iijima M, Yoshimoto N, Maturana AD, Ting K, Kuroda S, Niimi T. Oligomerization-induced conformational change in the C-terminal region of Nel-like molecule 1 (NELL1) protein is necessary for the efficient mediation of murine MC3T3-E1 cell adhesion and spreading. J Biol Chem 2014; 289:9781-94. [PMID: 24563467 DOI: 10.1074/jbc.m113.507020] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
NELL1 is a large oligomeric secretory glycoprotein that functions as an osteoinductive factor. NELL1 contains several conserved domains, has structural similarities to thrombospondin 1, and supports osteoblastic cell adhesion through integrins. To define the structural requirements for NELL1-mediated cell adhesion, we prepared a series of recombinant NELL1 proteins (intact, deleted, and cysteine-mutant) from a mammalian expression system and tested their activities. A deletion analysis demonstrated that the C-terminal cysteine-rich region of NELL1 is critical for the cell adhesion activity of NELL1. Reducing agent treatment decreased the cell adhesion activity of full-length NELL1 but not of its C-terminal fragments, suggesting that the intramolecular disulfide bonds within this region are not functionally necessary but that other disulfide linkages in the N-terminal region of NELL1 may be involved in cell adhesion activity. By replacing cysteine residues with serines around the coiled-coil domain of NELL1, which is responsible for oligomerization, we created a mutant NELL1 protein that was unable to form homo-oligomers, and this monomeric mutant showed substantially lower cell adhesion activity than intact NELL1. These results suggest that an oligomerization-induced conformational change in the C-terminal region of NELL1 is important for the efficient mediation of cell adhesion and spreading by NELL1.
Collapse
Affiliation(s)
- Yoko Nakamura
- From the Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan and
| | | | | | | | | | | | | | | | | |
Collapse
|