1
|
Siboni R, Sergheraert J, Thoraval L, Guillaume C, Gangloff SC, Ohl X, Braux J, Velard F. Optimisation and Validation of an Induced Membrane Technique Model to Assess Bone Regeneration in Rats. J Tissue Eng Regen Med 2025; 2025:7357277. [PMID: 40297017 PMCID: PMC12037239 DOI: 10.1155/term/7357277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 03/11/2025] [Indexed: 04/30/2025]
Abstract
Background: The induced membrane (IM) preclinical models have been described in small animals, but few studies have looked at bone regeneration achievement. The optimisation and validation of such a preclinical model, considering the results obtained after the use of biomaterials as a substitute for bone grafting, could lead to simplifying the surgical procedure and enhance the clinical results. Methods: An in vivo model of the IM technique was developed on the femur of Lewis rats after a 4-mm critical bone defect stabilised with an osteosynthesis plate. A first optimisation phase was performed by evaluating different osteotomy methods and two different osteosynthesis plate sizes. The efficiency of the model was evaluated by the failure rate obtained 6 weeks after the first operative time. Thereafter, bone regeneration was evaluated histologically and radiologically at 24 weeks to confirm the critical nature of the bone defect (negative control), the effectiveness of the IM with a syngeneic bone graft (positive control) and the possibility of using a biomaterial (GlassBone Noraker) in this model. Results: Sixty-three rats were included and underwent the first surgical step. Nineteen rats subsequently underwent the second surgical step. The results obtained led to select piezotomy as the best osteotomy technique and 1-mm-thick plates with 2.0-mm-diameter screws as osteosynthesis material. Twenty-four weeks after the second surgical step, solely the group with both surgical steps and a syngeneic bone graft showed complete ossification of the bone defect. In contrast, the group without a graft did not present a suitable ossification, which confirms the critical nature of the defect. IM produced an incomplete bone regeneration using GlassBone alone. Conclusions: A piezotome osteotomy with an osteosynthesis plate of sufficient stiffness is required for this two-stage bone regeneration model in rats. The 4-mm bone defect is critical for this model and suitable for biomaterial evaluation.
Collapse
Affiliation(s)
- Renaud Siboni
- Biomaterial and Inflammation in Bone Site Laboratory (EA 4691 Bios), University of Reims Champagne Ardenne, 51 Rue Cognacq Jay, Reims 51100, France
- Department of Orthopaedic Surgery, Reims University Hospital, Hôpital Maison Blanche, 45 Rue Cognacq-Jay, Reims 51092, France
- UFR Médecine, University of Reims Champagne Ardenne, 51 Rue Cognacq Jay, Reims 51100, France
| | - Johan Sergheraert
- Biomaterial and Inflammation in Bone Site Laboratory (EA 4691 Bios), University of Reims Champagne Ardenne, 51 Rue Cognacq Jay, Reims 51100, France
- Department of Dental Surgery, Reims University Hospital, Hôpital Maison Blanche, 45 Rue Cognacq-Jay, Reims 51092, France
| | - Lea Thoraval
- Biomaterial and Inflammation in Bone Site Laboratory (EA 4691 Bios), University of Reims Champagne Ardenne, 51 Rue Cognacq Jay, Reims 51100, France
| | - Christine Guillaume
- Biomaterial and Inflammation in Bone Site Laboratory (EA 4691 Bios), University of Reims Champagne Ardenne, 51 Rue Cognacq Jay, Reims 51100, France
| | - Sophie C. Gangloff
- Biomaterial and Inflammation in Bone Site Laboratory (EA 4691 Bios), University of Reims Champagne Ardenne, 51 Rue Cognacq Jay, Reims 51100, France
- UFR Pharmacy, University of Reims Champagne Ardenne, 51 Rue Cognacq Jay, Reims 51100, France
| | - Xavier Ohl
- Biomaterial and Inflammation in Bone Site Laboratory (EA 4691 Bios), University of Reims Champagne Ardenne, 51 Rue Cognacq Jay, Reims 51100, France
- Department of Orthopaedic Surgery, Reims University Hospital, Hôpital Maison Blanche, 45 Rue Cognacq-Jay, Reims 51092, France
| | - Julien Braux
- Biomaterial and Inflammation in Bone Site Laboratory (EA 4691 Bios), University of Reims Champagne Ardenne, 51 Rue Cognacq Jay, Reims 51100, France
- Department of Dental Surgery, Reims University Hospital, Hôpital Maison Blanche, 45 Rue Cognacq-Jay, Reims 51092, France
| | - Frédéric Velard
- Biomaterial and Inflammation in Bone Site Laboratory (EA 4691 Bios), University of Reims Champagne Ardenne, 51 Rue Cognacq Jay, Reims 51100, France
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Ehlen Q, Costello JP, Mirsky NA, Slavin BV, Parra M, Ptashnik A, Nayak VV, Coelho PG, Witek L. Treatment of Bone Defects and Nonunion via Novel Delivery Mechanisms, Growth Factors, and Stem Cells: A Review. ACS Biomater Sci Eng 2024; 10:7314-7336. [PMID: 39527574 PMCID: PMC11632667 DOI: 10.1021/acsbiomaterials.4c01279] [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: 07/11/2024] [Revised: 10/18/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Bone nonunion following a fracture represents a significant global healthcare challenge, with an overall incidence ranging between 2 and 10% of all fractures. The management of nonunion is not only financially prohibitive but often necessitates invasive surgical interventions. This comprehensive manuscript aims to provide an extensive review of the published literature involving growth factors, stem cells, and novel delivery mechanisms for the treatment of fracture nonunion. Key growth factors involved in bone healing have been extensively studied, including bone morphogenic protein (BMP), vascular endothelial growth factor (VEGF), and platelet-derived growth factor. This review includes both preclinical and clinical studies that evaluated the role of growth factors in acute and chronic nonunion. Overall, these studies revealed promising bridging and fracture union rates but also elucidated complications such as heterotopic ossification and inferior mechanical properties associated with chronic nonunion. Stem cells, particularly mesenchymal stem cells (MSCs), are an extensively studied topic in the treatment of nonunion. A literature search identified articles that demonstrated improved healing responses, osteogenic capacity, and vascularization of fractures due to the presence of MSCs. Furthermore, this review addresses novel mechanisms and materials being researched to deliver these growth factors and stem cells to nonunion sites, including natural/synthetic polymers and bioceramics. The specific mechanisms explored in this review include BMP-induced osteoblast differentiation, VEGF-mediated angiogenesis, and the role of MSCs in multilineage differentiation and paracrine signaling. While these therapeutic modalities exhibit substantial preclinical promise in treating fracture nonunion, there remains a need for further research, particularly in chronic nonunion and large animal models. This paper seeks to identify such translational hurdles which must be addressed in order to progress the aforementioned treatments from the lab to the clinical setting.
Collapse
Affiliation(s)
- Quinn
T. Ehlen
- University
of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Joseph P. Costello
- University
of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Nicholas A. Mirsky
- University
of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Blaire V. Slavin
- University
of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Marcelo Parra
- Center
of Excellence in Morphological and Surgical Studies (CEMyQ), Faculty
of Medicine, Universidad de La Frontera, Temuco 4811230, Chile
- Department
of Comprehensive Adult Dentistry, Faculty of Dentistry, Universidad de La Frontera, Temuco 4811230, Chile
| | - Albert Ptashnik
- Biomaterials
Division, NYU Dentistry, New York, New York 10010, United States
| | - Vasudev Vivekanand Nayak
- Department
of Biochemistry and Molecular Biology, University
of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Paulo G. Coelho
- Department
of Biochemistry and Molecular Biology, University
of Miami Miller School of Medicine, Miami, Florida 33136, United States
- Division
of Plastic Surgery, DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Lukasz Witek
- Biomaterials
Division, NYU Dentistry, New York, New York 10010, United States
- Department
of Biomedical Engineering, NYU Tandon School
of Engineering, Brooklyn, New York 11201, United States
- Hansjörg
Wyss Department of Plastic Surgery, NYU
Grossman School of Medicine, New
York, New York 10016, United States
| |
Collapse
|
5
|
Bidkhori M, Akbarzadeh M, Fahimfar N, Jahangiri M, Seddiq S, Larijani B, Nabipour I, Mohammad Amoli M, Panahi N, Dehghan A, Holakouie-Naieni K, Ostovar A. Neural EGFL like 1 as a novel gene for Trabecular Bone Score in older adults: The Bushehr Elderly Health (BEH) program. PLoS One 2024; 19:e0309401. [PMID: 39255297 PMCID: PMC11386414 DOI: 10.1371/journal.pone.0309401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/13/2024] [Indexed: 09/12/2024] Open
Abstract
Neural EGFL like 1 (NELL-1), is a secreted glycoprotein and stimulates osteogenic cell differentiation and bone mineralization. This study aimed to explore the relationship between NELL-1 and Trabecular Bone Score (TBS) as a novel tool for the evaluation of osteoporosis in an elderly population-based cohort study in Iran. A single-locus analysis was performed on TBS using data from 2,071 participants in the Bushehr Elderly Health (BEH) Program. The study investigated 376 independent single nucleotide polymorphisms (SNPs) within the NELL-1 on chromosome 11p15.1. The association between SNPs and the mean TBS L1 to L4 was analyzed through an additive model. Significant variants in the additive model (PFDR<0.05) were further examined within dominant, recessive, over-dominant, and co-dominant models. Multiple linear regression was employed to assess the relationship between the genetic risk score (GRS) derived from significant SNPs and TBS. Three SNPs within the NELL-1 showed a statistically significant association with TBS after adjusting for age and sex. The associations for rs1901945 (β = 0.013, PFDR = 0.0007), rs1584851 (β = -0.011, PFDR = 0.0003), and rs58028601 (β = 0.011, PFDR = 0.0003) were significant in the additive model. Additionally, significant results were observed for rs1901945 and rs58028601 in the dominant model (P<0.05). The GRS showed a statistically significant relationship with TBS, considering adjustments for age, sex, Body Mass Index, type 2 diabetes, and smoking (β = 0.077, P = 1.7×10-5). This study highlights the association of NELL-1 with TBS, underscoring its potential as a candidate for further research and personalized medicine concerning the impact of this gene on bone quality.
Collapse
Affiliation(s)
- Mohammad Bidkhori
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Osteoporosis Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Akbarzadeh
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Noushin Fahimfar
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Osteoporosis Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Jahangiri
- Department of Biostatistics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sahar Seddiq
- Osteoporosis Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Iraj Nabipour
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mahsa Mohammad Amoli
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular -Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Nekoo Panahi
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular -Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Dehghan
- Department of Biostatistics and Epidemiology, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, United Kingdom
| | - Kourosh Holakouie-Naieni
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Afshin Ostovar
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Osteoporosis Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Ni C, Liao W, Nie H, Ge R, Liu R, Zou X, Yuan Z, Yan F. Neural epidermal growth factor-like 1 protein (Nell-1) alleviates periodontal tissue destruction in periodontitis by regulating the ratio of M2/M1 macrophage phenotypes. Int Immunopharmacol 2024; 137:112522. [PMID: 38908089 DOI: 10.1016/j.intimp.2024.112522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 06/01/2024] [Accepted: 06/16/2024] [Indexed: 06/24/2024]
Abstract
BACKGROUND Periodontitis is a common oral disease with high prevalence worldwide. Neural epidermal growth factor-like 1 protein (Nell-1) has recently been reported to have anti-inflammation effects and may be a drug candidate for osteoarthritis. However, its immunotherapeutic effects in periodontitis remain unknown. Therefore, this study aimed to investigate the effects of Nell-1 on periodontitis in terms of macrophage polarization and analyze its possible underlying mechanism. METHODS A rat ligation-induced experimental periodontitis model was established and locally injected with Nell-1 (n = 6/group). Periodontal tissue destruction and macrophage polarization in vivo were analyzed using micro-CT, histology analysis, and western blot. Enzyme-linked immunosorbent assay was used to evaluate serum inflammatory cytokines. Then, the RAW 264.7 macrophage cells were treated with lipopolysaccharide (LPS), Nell-1, and the c-Jun N-terminal kinases (JNK) inhibitor (SP600125). RT-PCR, western blot, and flow cytometry were performed to further analyze the effect of Nell-1 on macrophage polarization and the underlying mechanism in vitro. RESULTS Local treatment with Nell-1 significantly alleviated the destruction of alveolar bone and fibers in periodontitis, and upregulated the ratio of M2/M1 macrophages in periodontal tissues (P < 0.05). In vitro, Nell-1 at the concentrations of 200 and 500 ng/mL could significantly inhibit the expression of M1-related inflammatory factors in LPS-stimulated macrophages, and increase the expression of M2-related markers, regulating the macrophage phenotype switch into M2 (P < 0.05). The mRNA of JNK and relative protein level of phospho-JNK/JNK were also upregulated by Nell-1 (P < 0.05). Additionally, the JNK inhibitor (SP600125) could reverse the effect of Nell-1 on macrophage polarization (P < 0.05). CONCLUSIONS Nell-1 could modulate the ratio of M2/M1 macrophages possibly through the JNK/MAPK signaling pathway, subsequently attenuating the inflammation and destruction of periodontal tissues caused by periodontitis.
Collapse
Affiliation(s)
- Can Ni
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing 210008, China
| | - Wenzheng Liao
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing 210008, China
| | - Hua Nie
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing 210008, China
| | - Ruiyang Ge
- School of Stomatology, Zunyi Medical University, Zunyi 563099, China
| | - Rong Liu
- School of Stomatology, Zunyi Medical University, Zunyi 563099, China
| | - Xihong Zou
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing 210008, China
| | - Zhiyao Yuan
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing 210008, China
| | - Fuhua Yan
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing 210008, China.
| |
Collapse
|
7
|
Bonartsev A, Voinova V, Volkov A, Muraev A, Boyko E, Venediktov A, Didenko N, Dolgalev A. Scaffolds Based on Poly(3-Hydroxybutyrate) and Its Copolymers for Bone Tissue Engineering (Review). Sovrem Tekhnologii Med 2022; 14:78-90. [PMID: 37181830 PMCID: PMC10171059 DOI: 10.17691/stm2022.14.5.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Indexed: 05/16/2023] Open
Abstract
Biodegradable and biocompatible polymers are actively used in tissue engineering to manufacture scaffolds. Biomedical properties of polymer scaffolds depend on the physical and chemical characteristics and biodegradation kinetics of the polymer material, 3D microstructure and topography of the scaffold surface, as well as availability of minerals, medicinal agents, and growth factors loaded into the scaffold. However, in addition to the above, the intrinsic biological activity of the polymer and its biodegradation products can also become evident. This review provides studies demonstrating that scaffolds made of poly(3-hydroxybutyrate) (PHB) and its copolymers have their own biological activity, and namely, osteoinductive properties. PHB can induce differentiation of mesenchymal stem cells in the osteogenic direction in vitro and stimulates bone tissue regeneration during the simulation of critical and non-critical bone defects in vivo.
Collapse
Affiliation(s)
- A.P. Bonartsev
- Associate Professor, Department of Bioengineering, Faculty of Biology; Lomonosov Moscow State University, 1–12 Leninskiye Gory, Moscow, 119234, Russia
| | - V.V. Voinova
- Senior Researcher, Department of Biochemistry, Faculty of Biology; Lomonosov Moscow State University, 1–12 Leninskiye Gory, Moscow, 119234, Russia
| | - A.V. Volkov
- Senior Researcher; N.N. Priorov National Medical Research Center of Traumatology and Orthopedics, 10 Priorova St., Moscow, 127299, Russia; Associate Professor, Department of Pathological Anatomy, Medical Institute; Peoples’ Friendship University of Russia, 6 Miklukho-Maklaya St., Moscow, 117198, Russia
| | - A.A. Muraev
- Professor, Department of Maxillofacial Surgery and Surgical Dentistry; Peoples’ Friendship University of Russia, 6 Miklukho-Maklaya St., Moscow, 117198, Russia
| | - E.M. Boyko
- Teacher, Essentuki Branch; Stavropol State Medical University, 310 Mira St., Stavropol, 355017, Russia
| | - A.A. Venediktov
- Director; Cardioplant LLC, 1B Tsentralnaya St., Bldg. 2, Penza, 440004, Russia
| | - N.N. Didenko
- Assistant, Department of Pathological Physiology; Stavropol State Medical University, 310 Mira St., Stavropol, 355017, Russia
| | - A.A. Dolgalev
- Associate Professor, Professor, Department of General and Pediatric Dentistry; Stavropol State Medical University, 310 Mira St., Stavropol, 355017, Russia; Head of the Center for Innovation and Technology Transfer of the Research and Innovation Association; Stavropol State Medical University, 310 Mira St., Stavropol, 355017, Russia
| |
Collapse
|
8
|
Qin Q, Gomez-Salazar M, Tower RJ, Chang L, Morris CD, McCarthy EF, Ting K, Zhang X, James AW. NELL1 Regulates the Matrisome to Promote Osteosarcoma Progression. Cancer Res 2022; 82:2734-2747. [PMID: 35700263 PMCID: PMC9357190 DOI: 10.1158/0008-5472.can-22-0732] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/22/2022] [Accepted: 06/08/2022] [Indexed: 02/05/2023]
Abstract
Sarcomas produce an abnormal extracellular matrix (ECM), which in turn provides instructive cues for cell growth and invasion. Neural EGF like-like molecule 1 (NELL1) is a secreted glycoprotein characterized by its nonneoplastic osteoinductive effects, yet it is highly expressed in skeletal sarcomas. Here, we show that genetic deletion of NELL1 markedly reduces invasive behavior across human osteosarcoma (OS) cell lines. NELL1 deletion resulted in reduced OS disease progression, inhibiting metastasis and improving survival in a xenograft mouse model. These observations were recapitulated with Nell1 conditional knockout in mouse models of p53/Rb-driven sarcomagenesis, which reduced tumor frequency and extended tumor-free survival. Transcriptomic and phosphoproteomic analyses demonstrated that NELL1 loss skews the expression of matricellular proteins associated with reduced FAK signaling. Culturing NELL1 knockout sarcoma cells on wild-type OS-enriched matricellular proteins reversed the phenotypic and signaling changes induced by NELL1 deficiency. In sarcoma patients, high expression of NELL1 correlated with decreased overall survival. These findings in mouse and human models suggest that NELL1 expression alters the sarcoma ECM, thereby modulating cellular invasive potential and prognosis. Disruption of NELL1 signaling may represent a novel therapeutic approach to short-circuit sarcoma disease progression. SIGNIFICANCE NELL1 modulates the sarcoma matrisome to promote tumor growth, invasion, and metastasis, identifying the matrix-associated protein as an orchestrator of cell-ECM interactions in sarcomagenesis and disease progression.
Collapse
Affiliation(s)
- Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205
| | | | - Robert J. Tower
- Department of Orthopaedics, Johns Hopkins University, Baltimore, MD 21205
| | - Leslie Chang
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205
| | - Carol D. Morris
- Department of Orthopaedics, Johns Hopkins University, Baltimore, MD 21205
| | | | - Kang Ting
- Forsyth Institute, Cambridge, MA 02142
| | - Xinli Zhang
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095
| | - Aaron W. James
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205,Corresponding Author: Aaron W. James, M.D., Ph.D., 720 Rutland Avenue, Room 524A, Baltimore, MD 21205, Phone: (410) 502-4143,
| |
Collapse
|
9
|
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
|
10
|
Duan C, Townley HE. Isolation of NELL 1 Aptamers for Rhabdomyosarcoma Targeting. Bioengineering (Basel) 2022; 9:bioengineering9040174. [PMID: 35447734 PMCID: PMC9032205 DOI: 10.3390/bioengineering9040174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/06/2022] [Accepted: 04/13/2022] [Indexed: 12/13/2022] Open
Abstract
NELL1 (Neural epidermal growth factor-like (EGFL)-like protein) is an important biomarker associated with tissue and bone development and regeneration. NELL1 upregulation has been linked with metastasis and negative prognosis in rhabdomyosarcoma (RMS). Furthermore, multiple recent studies have also shown the importance of NELL1 in inflammatory bowel disease and membranous nephropathy, amongst other diseases. In this study, several anti-NELL1 DNA aptamers were selected from a randomized ssDNA pool using a fluorescence-guided method and evaluated for their binding affinity and selectivity. Several other methods such as a metabolic assay and confocal microscopy were also applied for the evaluation of the selected aptamers. The top three candidates were evaluated further, and AptNCan3 was shown to have a binding affinity up to 959.2 nM. Selectivity was examined in the RH30 RMS cells that overexpressed NELL1. Both AptNCan2 and AptNCan3 could significantly suppress metabolic activity in RMS cells. AptNCan3 was found to locate on the cell membrane and also on intracellular vesicles, which matched the location of NELL1 shown by antibodies in previous research. These results indicate that the selected anti-NELL1 aptamer showed strong and highly specific binding to NELL1 and therefore has potential to be used for in vitro or in vivo studies and treatments.
Collapse
Affiliation(s)
- Chengchen Duan
- Nuffield Department of Women’s and Reproductive Health, Oxford University John Radcliffe Hospital, Oxford OX3 9DU, UK;
| | - Helen Elizabeth Townley
- Nuffield Department of Women’s and Reproductive Health, Oxford University John Radcliffe Hospital, Oxford OX3 9DU, UK;
- Department of Engineering Science, Oxford University, Oxford OX1 3PJ, UK
- Correspondence: ; Tel.: +44-1865-283792
| |
Collapse
|
11
|
METTL3-Mediated lncRNA m 6A Modification in the Osteogenic Differentiation of Human Adipose-Derived Stem Cells Induced by NEL-Like 1 Protein. Stem Cell Rev Rep 2021; 17:2276-2290. [PMID: 34505967 DOI: 10.1007/s12015-021-10245-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2021] [Indexed: 02/05/2023]
Abstract
OBJECTIVES This study aimed to explore the regulatory mechanism of methyltransferase3 (METTL3) -mediated long non-coding RNA (lncRNA) N6-methyladenosine (m6A) modification in the osteogenic differentiation of human adipose-derived stem cells (hASCs) induced by NEL-like 1 protein (NELL-1). MATERIALS AND METHODS Methylated RNA immunoprecipitation sequencing (MeRIP-seq) and high- throughput sequencing for RNA (RNA-seq) were performed on hASCs. Osteogenic ability was detected by alkaline phosphatase (ALP) staining, Alizarin Red S(ARS) staining, ALP quantification and Quantitative real-time polymerase chain reaction analysis (qRT-PCR). Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis predicted the osteogenesis-related pathways enriched for the lncRNAs and identified the target lncRNAs. After overexpression and knockdown of METTL3, methylated RNA immunoprecipitation-qPCR (MeRIP-qPCR) and qRT-PCR were used to detect the levels of m6A modification and the expression of the target lncRNA, and the binding of both was confirmed by RNA binding protein immunoprecipitation (RIP) assay. The effects of lncRNA and METTL3 on phosphorylation of the key proteins of the pathway were detected by western blot analysis. RESULTS In vitro experiments showed that METTL3 can promote osteogenic differentiation and that its expression level is upregulated. KEGG pathway analysis predicted that lncRNAs with differentially upregulated methylated peaks were enriched mostly in the mitogen-activated protein kinase (MAPK) signaling pathway, in which Serine/threonine protein kinase 3 (STK3) was the predicted target gene of the lncRNA RP11-44 N12.5. The m6A modification and expression of RP11-44 N12.5 were both regulated by METTL3. Subsequently, lncRNA RP11-44 N12.5 and METTL3 were found to regulate the phosphorylation levels of three key proteins in the MAPK signaling pathway, ERK, JNK and p38. CONCLUSIONS This study shows, for the first time, that METTL3 can activate the MAPK signaling pathway by regulating the m6A modification and expression of a lncRNA, thereby enhancing the osteogenic differentiation of hASCs.
Collapse
|
12
|
Klein C, Monet M, Barbier V, Vanlaeys A, Masquelet AC, Gouron R, Mentaverri R. The Masquelet technique: Current concepts, animal models, and perspectives. J Tissue Eng Regen Med 2020; 14:1349-1359. [PMID: 32621637 DOI: 10.1002/term.3097] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 06/02/2020] [Accepted: 06/15/2020] [Indexed: 12/20/2022]
Abstract
Bone reconstruction within a critical-sized defect remains a real challenge in orthopedic surgery. The Masquelet technique is an innovative, two-step therapeutic approach for bone reconstruction in which the placement of a poly (methylmethacrylate) spacer into the bone defect induces the neo-formation of a tissue called "induced membrane." This surgical technique has many advantages and is often preferred to a vascularized bone flap or Ilizarov's technique. Although the Masquelet technique has achieved high clinical success rates since its development by Alain-Charles Masquelet in the early 2000s, very little is known about how the process works, and few animal models of membrane induction have been developed. Our successful use of this technique in the clinic and our interest in the mechanisms of tissue regeneration (notably bone regeneration) prompted us to develop a surgical model of the Masquelet technique in rats. Here, we provide a comprehensive review of the literature on animal models of membrane induction, encompassing the defect site, the surgical procedure, and the histologic and osteogenic properties of the induced membrane. We also discuss the advantages and disadvantages of those models to facilitate efforts in characterizing the complex biological mechanisms that underlie membrane induction.
Collapse
Affiliation(s)
- Céline Klein
- Department of Pediatric Orthopedic Surgery, Amiens University Medical Center, Jules Verne University of Picardie, Amiens, France.,MP3CV-EA7517, CURS, miens University Medical Center, Jules Verne University of Picardie, Amiens, France
| | - Michael Monet
- MP3CV-EA7517, CURS, miens University Medical Center, Jules Verne University of Picardie, Amiens, France
| | - Vincent Barbier
- Department of Pediatric Orthopedic Surgery, Amiens University Medical Center, Jules Verne University of Picardie, Amiens, France.,MP3CV-EA7517, CURS, miens University Medical Center, Jules Verne University of Picardie, Amiens, France
| | - Alison Vanlaeys
- MP3CV-EA7517, CURS, miens University Medical Center, Jules Verne University of Picardie, Amiens, France
| | - Alain-Charles Masquelet
- Service de Chirurgie Orthopédique, Traumatologie et Chirurgie de la Main, Saint-Antoine Hospital, Paris, France
| | - Richard Gouron
- Department of Pediatric Orthopedic Surgery, Amiens University Medical Center, Jules Verne University of Picardie, Amiens, France.,MP3CV-EA7517, CURS, miens University Medical Center, Jules Verne University of Picardie, Amiens, France
| | - Romuald Mentaverri
- MP3CV-EA7517, CURS, miens University Medical Center, Jules Verne University of Picardie, Amiens, France.,Department of Biochemistry and Endocrine Biology, Amiens University Medical Center, Jules Verne University of Picardie, Amiens, France
| |
Collapse
|
13
|
Zakrzewski W, Dobrzynski M, Rybak Z, Szymonowicz M, Wiglusz RJ. Selected Nanomaterials' Application Enhanced with the Use of Stem Cells in Acceleration of Alveolar Bone Regeneration during Augmentation Process. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1216. [PMID: 32580409 PMCID: PMC7353104 DOI: 10.3390/nano10061216] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/14/2020] [Accepted: 06/16/2020] [Indexed: 01/15/2023]
Abstract
Regenerative properties are different in every human tissue. Nowadays, with the increasing popularity of dental implants, bone regenerative procedures called augmentations are sometimes crucial in order to perform a successful dental procedure. Tissue engineering allows for controlled growth of alveolar and periodontal tissues, with use of scaffolds, cells, and signalling molecules. By modulating the patient's tissues, it can positively influence poor integration and healing, resulting in repeated implant surgeries. Application of nanomaterials and stem cells in tissue regeneration is a newly developing field, with great potential for maxillofacial bony defects. Nanostructured scaffolds provide a closer structural support with natural bone, while stem cells allow bony tissue regeneration in places when a certain volume of bone is crucial to perform a successful implantation. Several types of selected nanomaterials and stem cells were discussed in this study. Their use has a high impact on the efficacy of the current and future procedures, which are still challenging for medicine. There are many factors that can influence the regenerative process, while its general complexity makes the whole process even harder to control. The aim of this study was to evaluate the effectiveness and advantage of both stem cells and nanomaterials in order to better understand their function in regeneration of bone tissue in oral cavity.
Collapse
Affiliation(s)
- Wojciech Zakrzewski
- Department of Experimental Surgery and Biomaterial Research, Wroclaw Medical University, Bujwida 44, 50-345 Wroclaw, Poland; (W.Z.); (Z.R.); (M.S.)
| | - Maciej Dobrzynski
- Department of Conservative Dentistry and Pedodontics, Wroclaw Medical University, Krakowska 26, 50-425 Wroclaw, Poland;
| | - Zbigniew Rybak
- Department of Experimental Surgery and Biomaterial Research, Wroclaw Medical University, Bujwida 44, 50-345 Wroclaw, Poland; (W.Z.); (Z.R.); (M.S.)
| | - Maria Szymonowicz
- Department of Experimental Surgery and Biomaterial Research, Wroclaw Medical University, Bujwida 44, 50-345 Wroclaw, Poland; (W.Z.); (Z.R.); (M.S.)
| | - Rafal J. Wiglusz
- Institute of Low Temperature and Structure Research, Polish Academy of Sciences, Okolna 2, 50-422 Wroclaw, Poland
| |
Collapse
|
14
|
Wang D, Gilbert JR, Zhang X, Zhao B, Ker DFE, Cooper GM. Calvarial Versus Long Bone: Implications for Tailoring Skeletal Tissue Engineering. TISSUE ENGINEERING PART B-REVIEWS 2019; 26:46-63. [PMID: 31588853 DOI: 10.1089/ten.teb.2018.0353] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tissue-engineered graft substitutes have shown great potential to treat large bone defects. While we usually assume that therapeutic approaches developed for appendicular bone healing could be similarly translated for application in craniofacial reconstruction and vice versa, this is not necessarily accurate. In addition to those more well-known healing-associated factors, such as age, lifestyle (e.g., nutrition and smoking), preexisting disease (e.g., diabetes), medication, and poor blood supply, the developmental origins and surrounding tissue of the wound sites can largely affect the fracture healing outcome as well as designed treatments. Therefore, the strategies developed for long bone fracture repair might not be suitable or directly applicable to skull bone repair. In this review, we discuss aspects of development, healing process, structure, and tissue engineering considerations between calvarial and long bones to assist in designing the tailored bone repair strategies. Impact Statement We summarized, in this review, the existing body of knowledge between long bone and calvarial bone with regard to their development and healing, tissue structure, and consideration of current tissue engineering strategies. By highlighting their similarities and differences, we propose that tailored tissue engineering strategies, such as scaffold features, growth factor usage, and the source of cells for tissue or region-specific bone repair, are necessary to ensure an optimized healing outcome.
Collapse
Affiliation(s)
- Dan Wang
- Department of Stomatology, Tenth People's Hospital of Tongji University, Shanghai, China.,Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - James R Gilbert
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania.,McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Xu Zhang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Bingkun Zhao
- Department of Stomatology, Tenth People's Hospital of Tongji University, Shanghai, China.,Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Dai Fei Elmer Ker
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Gregory M Cooper
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Oral Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
15
|
Li C, Zhang X, Zheng Z, Nguyen A, Ting K, Soo C. Nell-1 Is a Key Functional Modulator in Osteochondrogenesis and Beyond. J Dent Res 2019; 98:1458-1468. [PMID: 31610747 DOI: 10.1177/0022034519882000] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Neural EGFL-like 1 (Nell-1) is a well-studied osteogenic factor that has comparable osteogenic potency with the Food and Drug Administration-approved bone morphogenic protein 2 (BMP-2). In this review, which aims to summarize the advanced Nell-1 research in the past 10 y, we start with the correlation of structural and functional relevance of the Nell-1 protein with the identification of a specific receptor of Nell-1, contactin-associated protein-like 4 (Cntnap4), for osteogenesis. The indispensable role of Nell-1 in normal craniofacial and appendicular skeletal development and growth was also defined by using the newly developed tissue-specific Nell-1 knockout mouse lines in addition to the existing transgenic mouse models. With the achievements on Nell-1's osteogenic therapeutic evaluations from multiple preclinical animal models for local and systemic bone regeneration, the synergistic effect of Nell-1 with BMP-2 on osteogenesis, as well as the advantages of Nell-1 as an osteogenic protein with antiadipogenic, anti-inflammatory, and provascularized characteristics over BMP-2 in bone tissue engineering, is highlighted, which lays the groundwork for the clinical trial approval of Nell-1. At the molecular level, besides the mitogen-activated protein kinase (MAPK) signaling pathway, we emphasize the significant involvement of the Wnt/β-catenin pathway as well as the key regulatory molecules Runt-related transcription factor 2 (Runx2) in Nell-1-induced osteogenesis. In addition, the involvement of Nell-1 in chondrogenesis and its relevant pathologies have been revealed with the participation of the nuclear factor of activated T cells 1 (Nfatc1), Runx3, and Indian hedgehog (Ihh) signaling pathways, although the mechanistic insights of Nell-1's osteochondrogenic property will be continuously evolving. With this perspective, we elucidate some emerging and novel functional properties of Nell-1 in oral-dental and neural tissues that will be the frontiers of future Nell-1 studies beyond the context of bone and cartilage. As such, the therapeutic potential of Nell-1 continues to evolve and grow with continuous pursuit.
Collapse
Affiliation(s)
- C Li
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - X Zhang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Z Zheng
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - A Nguyen
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - K Ting
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - C Soo
- Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, Orthopaedic Hospital Research Center, University of California, Los Angeles, CA, USA
| |
Collapse
|
16
|
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
|
17
|
Wang C, Tanjaya J, Shen J, Lee S, Bisht B, Pan HC, Pang S, Zhang Y, Berthiaume EA, Chen E, Da Lio AL, Zhang X, Ting K, Guo S, Soo C. Peroxisome Proliferator-Activated Receptor-γ Knockdown Impairs Bone Morphogenetic Protein-2-Induced Critical-Size Bone Defect Repair. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:648-664. [PMID: 30593824 PMCID: PMC6412314 DOI: 10.1016/j.ajpath.2018.11.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 10/13/2018] [Accepted: 11/13/2018] [Indexed: 12/15/2022]
Abstract
The Food and Drug Administration-approved clinical dose (1.5 mg/mL) of bone morphogenetic protein-2 (BMP2) has been reported to induce significant adverse effects, including cyst-like adipose-infiltrated abnormal bone formation. These undesirable complications occur because of increased adipogenesis, at the expense of osteogenesis, through BMP2-mediated increases in the master regulatory gene for adipogenesis, peroxisome proliferator-activated receptor-γ (PPARγ). Inhibiting PPARγ during osteogenesis has been suggested to drive the differentiation of bone marrow stromal/stem cells toward an osteogenic, rather than an adipogenic, lineage. We demonstrate that knocking down PPARγ while concurrently administering BMP2 can reduce adipogenesis, but we found that it also impairs BMP2-induced osteogenesis and leads to bone nonunion in a mouse femoral segmental defect model. In addition, in vitro studies using the mouse bone marrow stromal cell line M2-10B4 and mouse primary bone marrow stromal cells confirmed that PPARγ knockdown inhibits BMP2-induced adipogenesis; attenuates BMP2-induced cell proliferation, migration, invasion, and osteogenesis; and escalates BMP2-induced cell apoptosis. More important, BMP receptor 2 and 1B expression was also significantly inhibited by the combined BMP2 and PPARγ knockdown treatment. These findings indicate that PPARγ is critical for BMP2-mediated osteogenesis during bone repair. Thus, uncoupling BMP2-mediated osteogenesis and adipogenesis using PPARγ inhibition to combat BMP2's adverse effects may not be feasible.
Collapse
Affiliation(s)
- Chenchao Wang
- Department of Plastic Surgery, First Hospital of China Medical University, Shenyang, People's Republic of China; Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California; Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, and Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Justine Tanjaya
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Jia Shen
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Soonchul Lee
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California; Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Bharti Bisht
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Hsin Chuan Pan
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Shen Pang
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Yulong Zhang
- Departments of Materials Science and Engineering, and Division of Advanced Prosthodontics, University of California, Los Angeles, Los Angeles, California
| | - Emily A Berthiaume
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Eric Chen
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Andrew L Da Lio
- Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, and Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Xinli Zhang
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Kang Ting
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Shu Guo
- Department of Plastic Surgery, First Hospital of China Medical University, Shenyang, People's Republic of China.
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, and Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
18
|
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
|
19
|
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
|
20
|
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
|
21
|
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
|
22
|
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
|
23
|
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
|
24
|
Shen J, James AW, Zhang X, Pang S, Zara JN, Asatrian G, Chiang M, Lee M, Khadarian K, Nguyen A, Lee KS, Siu RK, Tetradis S, Ting K, Soo C. Novel Wnt Regulator NEL-Like Molecule-1 Antagonizes Adipogenesis and Augments Osteogenesis Induced by Bone Morphogenetic Protein 2. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:419-34. [PMID: 26772960 DOI: 10.1016/j.ajpath.2015.10.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 09/23/2015] [Accepted: 10/16/2015] [Indexed: 01/28/2023]
Abstract
The differentiation factor NEL-like molecule-1 (NELL-1) has been reported as osteoinductive in multiple in vivo preclinical models. Bone morphogenetic protein (BMP)-2 is used clinically for skeletal repair, but in vivo administration can induce abnormal, adipose-filled, poor-quality bone. We demonstrate that NELL-1 combined with BMP2 significantly optimizes osteogenesis in a rodent femoral segmental defect model by minimizing the formation of BMP2-induced adipose-filled cystlike bone. In vitro studies using the mouse bone marrow stromal cell line M2-10B4 and human primary bone marrow stromal cells have confirmed that NELL-1 enhances BMP2-induced osteogenesis and inhibits BMP2-induced adipogenesis. Importantly, the ability of NELL-1 to direct BMP2-treated cells toward osteogenesis and away from adipogenesis requires intact canonical Wnt signaling. Overall, these studies establish the feasibility of combining NELL-1 with BMP2 to improve clinical bone regeneration and provide mechanistic insight into canonical Wnt pathway activity during NELL-1 and BMP2 osteogenesis. The novel abilities of NELL-1 to stimulate Wnt signaling and to repress adipogenesis may highlight new treatment approaches for bone loss in osteoporosis.
Collapse
Affiliation(s)
- Jia Shen
- Division of Growth and Development and Section of Orthodontics, UCLA School of Dentistry, Los Angeles, California; UCLA Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery and Orthopaedic Hospital Research Center at UCLA, Los Angeles, California
| | - Aaron W James
- Division of Growth and Development and Section of Orthodontics, UCLA School of Dentistry, Los Angeles, California; UCLA Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery and Orthopaedic Hospital Research Center at UCLA, Los Angeles, California; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, California
| | - Xinli Zhang
- Division of Growth and Development and Section of Orthodontics, UCLA School of Dentistry, Los Angeles, California; UCLA Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery and Orthopaedic Hospital Research Center at UCLA, Los Angeles, California
| | - Shen Pang
- UCLA Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery and Orthopaedic Hospital Research Center at UCLA, Los Angeles, California
| | - Janette N Zara
- Division of Growth and Development and Section of Orthodontics, UCLA School of Dentistry, Los Angeles, California; UCLA Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery and Orthopaedic Hospital Research Center at UCLA, Los Angeles, California
| | - Greg Asatrian
- Division of Growth and Development and Section of Orthodontics, UCLA School of Dentistry, Los Angeles, California
| | - Michael Chiang
- Division of Growth and Development and Section of Orthodontics, UCLA School of Dentistry, Los Angeles, California
| | - Min Lee
- Division of Advanced Prosthodontics, UCLA School of Dentistry, Los Angeles, California
| | - Kevork Khadarian
- UCLA Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery and Orthopaedic Hospital Research Center at UCLA, Los Angeles, California
| | - Alan Nguyen
- Division of Growth and Development and Section of Orthodontics, UCLA School of Dentistry, Los Angeles, California
| | - Kevin S Lee
- Division of Growth and Development and Section of Orthodontics, UCLA School of Dentistry, Los Angeles, California
| | - Ronald K Siu
- Division of Growth and Development and Section of Orthodontics, UCLA School of Dentistry, Los Angeles, California
| | - Sotirios Tetradis
- Division of Diagnostic and Surgical Sciences, UCLA School of Dentistry, Los Angeles, California
| | - Kang Ting
- Division of Growth and Development and Section of Orthodontics, UCLA School of Dentistry, Los Angeles, California.
| | - Chia Soo
- UCLA Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery and Orthopaedic Hospital Research Center at UCLA, Los Angeles, California.
| |
Collapse
|
25
|
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
|
26
|
A Novel Rat Model of Intramedullary Tibia Fracture Fixation Using Polyetheretherketone Threaded Rod. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2015; 3:e417. [PMID: 26180718 PMCID: PMC4494487 DOI: 10.1097/gox.0000000000000386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 04/15/2015] [Indexed: 12/15/2022]
Abstract
Supplemental Digital Content is available in the text. Background: The rat fracture fixation models have been widely adopted, but current implant designs suffer from operational difficulty, massive soft-tissue dissection, and radiological intervention. The authors developed a new tibia fracture-healing model using minor invasive intramedullary fixations with polyetheretherketone (PEEK) threaded rods, which have excellent x-ray translucency and no magnetic resonance artifact. Methods: Tibia fractures of 6 adult male Sprague-Dawley rats were fixed with intramedullary PEEK threaded rods. X-ray examination was performed at 0, 4, and 8 weeks postoperatively. Histological analysis was conducted via hematoxylin-eosin staining of nondecalcified tissue sections. Results: Radiological fracture healing was observed at 8 weeks postoperatively. Histology demonstrated fracture gap bridging and bone ingrowth adjacent to PEEK. Conclusion: This innovative model is simple and effective, providing a new selection in future biomedical research.
Collapse
|
27
|
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
|
28
|
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
|
29
|
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
|
30
|
Sato K, Watanabe Y, Harada N, Abe S, Matsushita T, Yamanaka K, Kaneko T, Sakai Y. Establishment of Reproducible, Critical-Sized, Femoral Segmental Bone Defects in Rats. Tissue Eng Part C Methods 2014; 20:1037-41. [DOI: 10.1089/ten.tec.2013.0612] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Kenji Sato
- Department of Orthopaedic Surgery, Teikyo University School of Medicine, Tokyo, Japan
| | - Yoshinobu Watanabe
- Department of Orthopaedic Surgery, Teikyo University School of Medicine, Tokyo, Japan
| | - Noriko Harada
- Department of Orthopaedic Surgery, Teikyo University School of Medicine, Tokyo, Japan
| | - Satoshi Abe
- Department of Orthopaedic Surgery, Teikyo University School of Medicine, Tokyo, Japan
| | - Takashi Matsushita
- Department of Orthopaedic Surgery, Teikyo University School of Medicine, Tokyo, Japan
| | - Katsuyuki Yamanaka
- Research and Development Department, GC Corporation, Itabashi, Tokyo, Japan
| | - Tadashi Kaneko
- Research and Development Department, GC Corporation, Itabashi, Tokyo, Japan
| | - Yuhiro Sakai
- Research and Development Department, GC Corporation, Itabashi, Tokyo, Japan
| |
Collapse
|
31
|
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
|
32
|
Current trends in bone tissue engineering. BIOMED RESEARCH INTERNATIONAL 2014; 2014:865270. [PMID: 24804256 PMCID: PMC3997160 DOI: 10.1155/2014/865270] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 03/09/2014] [Indexed: 01/20/2023]
Abstract
The development of tissue engineering and regeneration constitutes a new platform for translational medical research. Effective therapies for bone engineering typically employ the coordinated manipulation of cells, biologically active signaling molecules, and biomimetic, biodegradable scaffolds. Bone tissue engineering has become increasingly dependent on the merging of innovations from each of these fields, as they continue to evolve independently. This foreword will highlight some of the most recent advances in bone tissue engineering and regeneration, emphasizing the interconnected fields of stem cell biology, cell signaling biology, and biomaterial research. These include, for example, novel methods for mesenchymal stem cell purification, new methods of Wnt signaling pathway manipulation, and cutting edge computer assisted nanoscale design of bone scaffold materials. In the following
special issue, we sought to incorporate these diverse areas of emphasis in order to reflect current trends in the field.
Collapse
|
33
|
Turner NJ, Londono R, Dearth CL, Culiat CT, Badylak SF. Human NELL1 protein augments constructive tissue remodeling with biologic scaffolds. Cells Tissues Organs 2013; 198:249-65. [PMID: 24335144 DOI: 10.1159/000356491] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2013] [Indexed: 11/19/2022] Open
Abstract
Biologic scaffolds composed of extracellular matrix (ECM) derived from decellularized tissues effectively reprogram key stages of the mammalian response to injury, altering the wound microenvironment from one that promotes scar tissue formation to one that stimulates constructive and functional tissue remodeling. In contrast, engineered scaffolds, composed of purified ECM components such as collagen, lack the complex ultrastructure and composition of intact ECM and may promote wound healing but lack factors that facilitate constructive and functional tissue remodeling. The objective of the present study was to test the hypothesis that addition of NELL1, a signaling protein that controls cell growth and differentiation, enhances the constructive tissue remodeling of a purified collagen scaffold. An abdominal wall defect model in the rat of 1.5-cm(2) partial thickness was used to compare the constructive remodeling of a bovine type I collagen scaffold to a biologic scaffold derived from small intestinal submucosa (SIS)-ECM with and without augmentation with 17 μg NELL1 protein. Samples were evaluated histologically at 14 days and 4 months. The contractile response of the defect site was also evaluated at 4 months. Addition of NELL1 protein improved the constructive remodeling of collagen scaffolds but not SIS-ECM scaffolds. Results showed an increase in the contractile force of the remodeled skeletal muscle and a fast:slow muscle composition similar to native tissue in the collagen-treated group. The already robust remodeling response to SIS-ECM was not enhanced by NELL1 at the dose tested. These findings suggest that NELL1 protein does contribute to the enhanced constructive remodeling of skeletal muscle.
Collapse
Affiliation(s)
- Neill J Turner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pa., USA
| | | | | | | | | |
Collapse
|
34
|
Rose L, Uludağ H. Realizing the potential of gene-based molecular therapies in bone repair. J Bone Miner Res 2013; 28:2245-62. [PMID: 23553878 DOI: 10.1002/jbmr.1944] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 03/13/2013] [Accepted: 03/19/2013] [Indexed: 12/17/2022]
Abstract
A better understanding of osteogenesis at genetic and biochemical levels is yielding new molecular entities that can modulate bone regeneration and potentially act as novel therapies in a clinical setting. These new entities are motivating alternative approaches for bone repair by utilizing DNA-derived expression systems, as well as RNA-based regulatory molecules controlling the fate of cells involved in osteogenesis. These sophisticated mediators of osteogenesis, however, pose unique delivery challenges that are not obvious in deployment of conventional therapeutic agents. Viral and nonviral delivery systems are actively pursued in preclinical animal models to realize the potential of the gene-based medicines. This article will summarize promising bone-inducing molecular agents on the horizon as well as provide a critical review of delivery systems employed for their administration. Special attention was paid to synthetic (nonviral) delivery systems because they are more likely to be adopted for clinical testing because of safety considerations. We present a comparative analysis of dose-response relationships, as well as pharmacokinetic and pharmacodynamic features of various approaches, with the purpose of clearly defining the current frontier in the field. We conclude with the authors' perspective on the future of gene-based therapy of bone defects, articulating promising research avenues to advance the field of clinical bone repair.
Collapse
Affiliation(s)
- Laura Rose
- Department of Biomedical Engineering, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | | |
Collapse
|
35
|
Shen J, James AW, Zara JN, Asatrian G, Khadarian K, Zhang JB, Ho S, Kim HJ, Ting K, Soo C. BMP2-induced inflammation can be suppressed by the osteoinductive growth factor NELL-1. Tissue Eng Part A 2013; 19:2390-401. [PMID: 23758588 DOI: 10.1089/ten.tea.2012.0519] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Bone-morphogenetic protein 2 (BMP2) is currently the only Food and Drug Administration-approved osteoinductive growth factor used in clinical settings for bone regeneration and repair. However, the use of BMP2 is encumbered by numerous clinical complications, including postoperative inflammation and life-threatening cervical swelling. Thus, methods to prevent BMP2-induced inflammation would have far-reaching clinical implications toward improving current BMP2-based methods for bone regeneration. For the first time, we investigate the potential role of the growth factor Nel-like molecule-1 (NELL-1) in inhibiting BMP2-induced inflammation. Adult rats underwent a femoral bone onlay procedure, treated with either BMP2 protein (4 mg/mL), NELL-1 protein (4 mg/mL), or both proteins combined. Animals were evaluated at 3, 7, and 14 days postoperatively by histology, histomorphometry, immunohistochemistry, and real-time PCR for markers of inflammation (TNFα, IL6). The relative levels of TNFα and IL6 in serum were also detected by ELISA. The mechanism for NELL-1's anti-inflammatory effect was further assessed through examining inflammatory markers and generation of reactive oxygen species (ROS) in the mouse embryonic fibroblast NIH3T3 cells. BMP2 significantly induced local inflammation, including an early and pronounced polymorphonuclear cell infiltration accompanied by increased expression of TNFα and IL6. Treatment with NELL-1 alone elicited no significant inflammatory response. However, NELL-1 significantly attenuated BMP2-induced inflammation by all markers and at all timepoints. These local findings were also confirmed using systemic serum inflammatory biomarkers (TNFα, IL6). In each case, NELL-1 fully reversed BMP2-induced systemic inflammation. Lastly, our findings were recapitulated in vitro, where NELL-1 suppressed BMP2 induced expression of inflammatory markers, as well as NF-κB transcriptional activity and generation of ROS. BMP2-induced inflammation is a serious public health concern with potentially life-threatening complications. In the present study, we observed that the growth factor, NELL-1, significantly attenuates or completely reverses BMP2-induced inflammation. The mechanisms of NELL-1's anti-inflammatory effect are only partially elucidated, and may include reduction of NF-κB transcriptional activity or ROS generation.
Collapse
Affiliation(s)
- Jia Shen
- 1 Division of Associated Clinical Specialties, Section of Orthodontics, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Yuan W, James AW, Asatrian G, Shen J, Zara JN, Tian HJ, Siu RK, Zhang X, Wang JC, Dong J. NELL-1 based demineralized bone graft promotes rat spine fusion as compared to commercially available BMP-2 product. J Orthop Sci 2013; 18:646-57. [PMID: 23686083 DOI: 10.1007/s00776-013-0390-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Accepted: 03/21/2013] [Indexed: 11/26/2022]
Abstract
BACKGROUND Spinal fusion is among the most commonly performed orthopaedic procedures. Unfortunately, current treatments such as autologous bone grafting or recombinant proteins (BMP-2) have numerous clinical shortcomings. Here, we directly compare the efficacy of NELL-1, a novel osteoinductive growth factor, to two currently available treatments, (1) recombinant BMP-2 and (2) iliac crest bone grafting, in a spinal fusion model. METHODS Twenty-six skeletally mature athymic rats underwent posterolateral spine fusion of L4/L5 vertebrae. Treatment groups included NELL-1 (10 and 50 μg) in a demineralized bone matrix (DBX), as compared to BMP-2 (90 μg) in an absorbable collagen sponge (ACS) or morselized iliac crest bone. Scaffolds without recombinant protein were used as controls. Animals were sacrificed at 4 weeks post-operative and fusion was assessed by manual palpation, radiography [high-resolution X-ray, micro-computed tomography (microCT)], histology (hematoxylin and eosin, Masson's trichrome) and immunohistochemistry (osteocalcin). RESULTS Results showed 100 % fusion in all NELL-1- and BMP-2-treated samples. In contrast, lower rates of fusion were observed in scaffold-only and bone graft treatment groups. MicroCT scans revealed radiographic evidence of fusion among spines treated with NELL-1. Bone bridging was also observed with BMP-2 treatment, but was accompanied by inner radiolucency, suggesting cyst-like bone formation. Histologically, NELL-1-treated grafts showed increased bone formation, endochondral ossification and vascularization. Although BMP-2 treated grafts exhibited increased bone formation and angiogenesis, numerous adipocytes were also observed. CONCLUSION NELL-1-based bone grafts are comparable to BMP-2 + ACS in spinal fusion efficacy. Histological differences were observed however, including robust endochondral ossification with NELL-1 treatment as compared to lipid-filled bone with BMP-2 treatment. These findings suggest NELL-1 based bone grafts show promise for future efforts in skeletal tissue engineering.
Collapse
Affiliation(s)
- Wei Yuan
- Department of Orthopaedic Surgery, Zhongshan Hospital, FudanUniversity, 180 Fenglin Road, Shanghai 200032, China.e-mail:
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Regenerative medicine is a rapidly evolving multidisciplinary, translational research enterprise whose explicit purpose is to advance technologies for the repair and replacement of damaged cells, tissues, and organs. Scientific progress in the field has been steady and expectations for its robust clinical application continue to rise. The major thesis of this review is that the pharmacological sciences will contribute critically to the accelerated translational progress and clinical utility of regenerative medicine technologies. In 2007, we coined the phrase "regenerative pharmacology" to describe the enormous possibilities that could occur at the interface between pharmacology, regenerative medicine, and tissue engineering. The operational definition of regenerative pharmacology is "the application of pharmacological sciences to accelerate, optimize, and characterize (either in vitro or in vivo) the development, maturation, and function of bioengineered and regenerating tissues." As such, regenerative pharmacology seeks to cure disease through restoration of tissue/organ function. This strategy is distinct from standard pharmacotherapy, which is often limited to the amelioration of symptoms. Our goal here is to get pharmacologists more involved in this field of research by exposing them to the tools, opportunities, challenges, and interdisciplinary expertise that will be required to ensure awareness and galvanize involvement. To this end, we illustrate ways in which the pharmacological sciences can drive future innovations in regenerative medicine and tissue engineering and thus help to revolutionize the discovery of curative therapeutics. Hopefully, the broad foundational knowledge provided herein will spark sustained conversations among experts in diverse fields of scientific research to the benefit of all.
Collapse
Affiliation(s)
- George J Christ
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA.
| | | | | | | |
Collapse
|
38
|
Askarinam A, James AW, Zara JN, Goyal R, Corselli M, Pan A, Liang P, Chang L, Rackohn T, Stoker D, Zhang X, Ting K, Péault B, Soo C. Human perivascular stem cells show enhanced osteogenesis and vasculogenesis with Nel-like molecule I protein. Tissue Eng Part A 2013; 19:1386-97. [PMID: 23406369 PMCID: PMC3638559 DOI: 10.1089/ten.tea.2012.0367] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 01/16/2013] [Indexed: 12/31/2022] Open
Abstract
An ideal mesenchymal stem cell (MSC) source for bone tissue engineering has yet to be identified. Such an MSC population would be easily harvested in abundance, with minimal morbidity and with high purity. Our laboratories have identified perivascular stem cells (PSCs) as a candidate cell source. PSCs are readily isolatable through fluorescent-activated cell sorting from adipose tissue and have been previously shown to be indistinguishable from MSCs in the phenotype and differentiation potential. PSCs consist of two distinct cell populations: (1) pericytes (CD146+, CD34-, and CD45-), which surround capillaries and microvessels, and (2) adventitial cells (CD146-, CD34+, and CD45-), found within the tunica adventitia of large arteries and veins. We previously demonstrated the osteogenic potential of pericytes by examining pericytes derived from the human fetal pancreas, and illustrated their in vivo trophic and angiogenic effects. In the present study, we used an intramuscular ectopic bone model to develop the translational potential of our original findings using PSCs (as a combination of pericytes and adventitial cells) from human white adipose tissue. We evaluated human PSC (hPSC)-mediated bone formation and vascularization in vivo. We also examined the effects of hPSCs when combined with the novel craniosynostosis-associated protein, Nel-like molecule I (NELL-1). Implants consisting of the demineralized bone matrix putty combined with NELL-1 (3 μg/μL), hPSC (2.5×10(5) cells), or hPSC+NELL-1, were inserted in the bicep femoris of SCID mice. Bone growth was evaluated using microcomputed tomography, histology, and immunohistochemistry over 4 weeks. Results demonstrated the osteogenic potential of hPSCs and the additive effect of hPSC+NELL-1 on bone formation and vasculogenesis. Comparable osteogenesis was observed with NELL-1 as compared to the more commonly used bone morphogenetic protein-2. Next, hPSCs induced greater implant vascularization than the unsorted stromal vascular fraction from patient-matched samples. Finally, we observed an additive effect on implant vascularization with hPSC+NELL-1 by histomorphometry and immunohistochemistry, accompanied by in vitro elaboration of vasculogenic growth factors. These findings hold significant implications for the cell/protein combination therapy hPSC+NELL-1 in the development of strategies for vascularized bone regeneration.
Collapse
Affiliation(s)
- Asal Askarinam
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, California
| | - Aaron W. James
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, California
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Janette N. Zara
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Raghav Goyal
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, California
| | - Mirko Corselli
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Angel Pan
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, California
| | - Pei Liang
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Le Chang
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, California
| | - Todd Rackohn
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, California
| | - David Stoker
- Division of Plastic and Reconstructive Surgery, University of Southern California, Los Angeles, California
| | - Xinli Zhang
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, California
| | - Kang Ting
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA, Los Angeles, California
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Bruno Péault
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Chia Soo
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
39
|
DeConde AS, Sidell D, Lee M, Bezouglaia O, Low K, Elashoff D, Grogan T, Tetradis S, Aghaloo T, St John M. Bone morphogenetic protein-2-impregnated biomimetic scaffolds successfully induce bone healing in a marginal mandibular defect. Laryngoscope 2013; 123:1149-55. [PMID: 23553490 DOI: 10.1002/lary.23782] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 09/23/2012] [Accepted: 09/18/2012] [Indexed: 11/09/2022]
Abstract
OBJECTIVES/HYPOTHESIS To test the osteoregenerative potential and dosing of bone morphogenetic protein-2 (BMP-2)-impregnated biomimetic scaffolds in a rat model of a mandibular defect. STUDY DESIGN Prospective study using an animal model. METHODS Varied doses of BMP-2 (0.5, 1, 0.5, 0.5 in microspheres, 5, and 15 μg) were absorbed onto a biomimetic scaffold. Scaffolds were then implanted into marginal mandibular defects in rats. Blank scaffolds and unfilled defects were used as negative controls. Two months postoperatively, bone healing was analyzed with microcomputerized tomography (microCT). RESULTS MicroCT analysis demonstrated that all doses of BMP-2 induced successful healing of marginal mandibular defects in a rat mandible. Increasing doses of BMP-2 on the scaffolds produced increased tissue healing, with 15 μg demonstrating significantly more healing than all other dosing (P < .01). CONCLUSIONS BMP-2-impregnated biomimetic scaffolds successfully induce bone healing in a marginal mandibular defect in the rat. Percentage healing of defect, percentage of bone within healed tissue, and total bone volume are all a function of BMP-2 dosing. There appears to be an optimal dose of 5 μg beyond which there is no increase in bone volume. LEVEL OF EVIDENCE NA.
Collapse
Affiliation(s)
- Adam S DeConde
- Department of Head and Neck Surgery, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, California, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
James AW. Review of Signaling Pathways Governing MSC Osteogenic and Adipogenic Differentiation. SCIENTIFICA 2013; 2013:684736. [PMID: 24416618 PMCID: PMC3874981 DOI: 10.1155/2013/684736] [Citation(s) in RCA: 323] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 11/21/2013] [Indexed: 05/07/2023]
Abstract
Mesenchymal stem cells (MSC) are multipotent cells, functioning as precursors to a variety of cell types including adipocytes, osteoblasts, and chondrocytes. Between osteogenic and adipogenic lineage commitment and differentiation, a theoretical inverse relationship exists, such that differentiation towards an osteoblast phenotype occurs at the expense of an adipocytic phenotype. This balance is regulated by numerous, intersecting signaling pathways that converge on the regulation of two main transcription factors: peroxisome proliferator-activated receptor- γ (PPAR γ ) and Runt-related transcription factor 2 (Runx2). These two transcription factors, PPAR γ and Runx2, are generally regarded as the master regulators of adipogenesis and osteogenesis. This review will summarize signaling pathways that govern MSC fate towards osteogenic or adipocytic differentiation. A number of signaling pathways follow the inverse balance between osteogenic and adipogenic differentiation and are generally proosteogenic/antiadipogenic stimuli. These include β -catenin dependent Wnt signaling, Hedgehog signaling, and NELL-1 signaling. However, other signaling pathways exhibit more context-dependent effects on adipogenic and osteogenic differentiation. These include bone morphogenic protein (BMP) signaling and insulin growth factor (IGF) signaling, which display both proosteogenic and proadipogenic effects. In summary, understanding those factors that govern osteogenic versus adipogenic MSC differentiation has significant implications in diverse areas of human health, from obesity to osteoporosis to regenerative medicine.
Collapse
Affiliation(s)
- Aaron W. James
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, 10833 Le Conte Avenue, CHS A3-251, Los Angeles, CA 90077, USA
- *Aaron W. James:
| |
Collapse
|
41
|
The role of vertebrate models in understanding craniosynostosis. Childs Nerv Syst 2012; 28:1471-81. [PMID: 22872264 DOI: 10.1007/s00381-012-1844-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 06/13/2012] [Indexed: 01/10/2023]
Abstract
BACKGROUND Craniosynostosis (CS), the premature fusion of cranial sutures, is a relatively common pediatric anomaly, occurring in isolation or as part of a syndrome. A growing number of genes with pathologic mutations have been identified for syndromic and nonsyndromic CS. The study of human sutural material obtained post-operatively is not sufficient to understand the etiology of CS, for which animal models are indispensable. DISCUSSION The similarity of the human and murine calvarial structure, our knowledge of mouse genetics and biology, and ability to manipulate the mouse genome make the mouse the most valuable model organism for CS research. A variety of mouse mutants are available that model specific human CS mutations or have CS phenotypes. These allow characterization of the biochemical and morphological events, often embryonic, which precede suture fusion. Other vertebrate organisms have less functional genetic utility than mice, but the rat, rabbit, chick, zebrafish, and frog provide alternative systems in which to validate or contrast molecular functions relevant to CS.
Collapse
|
42
|
NELL-1-dependent mineralisation of Saos-2 human osteosarcoma cells is mediated via c-Jun N-terminal kinase pathway activation. INTERNATIONAL ORTHOPAEDICS 2012; 36:2181-7. [PMID: 22797704 DOI: 10.1007/s00264-012-1590-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 05/22/2012] [Indexed: 12/22/2022]
Abstract
PURPOSE NELL-1 is a novel osteoinductive growth factor that has shown promising results for the regeneration of bone. Moreover, NELL-1 has been used successfully in bone regeneration in the axial, appendicular and calvarial skeleton of both small and large animal models. Despite increasing evidence of NELL-1 efficacy and future usefulness as an alternative to traditional bone graft substitutes, much has yet to be understood regarding the mechanisms of action of this novel protein. The activation of the mitogen-activated protein kinase (MAPK) pathway has been well studied in the setting of growth factor-mediated changes in osteogenic differentiation. METHODS In this study, we provide evidence of the involvement of MAPK signalling pathways in NELL-1-induced terminal osteogenic differentiation of Saos-2 human osteosarcoma cells. Activation of extracellular signal-regulated kinase (ERK1/2), P38 and c-Jun N-terminal kinase (JNK) pathways were screened with MAPK signalling protein array after recombinant human (rh)NELL-1 treatment. Next, the mineralisation and intracellular phosphate levels after rhNELL-1 stimulation were assessed in the presence or absence of specific MAPK inhibitors. RESULTS Results showed that rhNELL-1 predominantly increased JNK pathway activation. Moreover, the specific JNK inhibitor SP600125 blocked rhNELL-1-induced mineralisation and intracellular phosphate accumulation, whereas ERK1/2 and P38 inhibitors showed no effect. CONCLUSIONS Thus, activation of the JNK pathway is necessary to mediate terminal osteogenic differentiation of Saos-2 osteosarcoma cells by rhNELL-1. Future studies will extend these in vitro mechanisms to the in vivo effects of NELL-1 in dealing with orthopaedic defects caused by skeletal malignancies or other aetiologies.
Collapse
|
43
|
James AW, Zara JN, Zhang X, Askarinam A, Goyal R, Chiang M, Yuan W, Chang L, Corselli M, Shen J, Pang S, Stoker D, Wu B, Ting K, Péault B, Soo C. Perivascular stem cells: a prospectively purified mesenchymal stem cell population for bone tissue engineering. Stem Cells Transl Med 2012. [PMID: 23197855 DOI: 10.5966/sctm.2012-0002] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Adipose tissue is an ideal source of mesenchymal stem cells for bone tissue engineering: it is largely dispensable and readily accessible with minimal morbidity. However, the stromal vascular fraction (SVF) of adipose tissue is a heterogeneous cell population, which leads to unreliable bone formation. In the present study, we prospectively purified human perivascular stem cells (PSCs) from adipose tissue and compared their bone-forming capacity with that of traditionally derived SVF. PSCs are a population (sorted by fluorescence-activated cell sorting) of pericytes (CD146+CD34-CD45-) and adventitial cells (CD146-CD34+CD45-), each of which we have previously reported to have properties of mesenchymal stem cells. Here, we found that PSCs underwent osteogenic differentiation in vitro and formed bone after intramuscular implantation without the need for predifferentiation. We next sought to optimize PSCs for in vivo bone formation, adopting a demineralized bone matrix for osteoinduction and tricalcium phosphate particle formulation for protein release. Patient-matched, purified PSCs formed significantly more bone in comparison with traditionally derived SVF by all parameters. Recombinant bone morphogenetic protein 2 increased in vivo bone formation but with a massive adipogenic response. In contrast, recombinant Nel-like molecule 1 (NELL-1; a novel osteoinductive growth factor) selectively enhanced bone formation. These studies suggest that adipose-derived human PSCs are a new cell source for future efforts in skeletal regenerative medicine. Moreover, PSCs are a stem cell-based therapeutic that is readily approvable by the U.S. Food and Drug Administration, with potentially increased safety, purity, identity, potency, and efficacy. Finally, NELL-1 is a candidate growth factor able to induce human PSC osteogenesis.
Collapse
Affiliation(s)
- Aaron W James
- Dental and Craniofacial Research Institute, University of California, Los Angeles, USA. 900950-1579, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
James AW, Zara JN, Corselli M, Chiang M, Yuan W, Nguyen V, Askarinam A, Goyal R, Siu RK, Scott V, Lee M, Ting K, Péault B, Soo C. Use of human perivascular stem cells for bone regeneration. J Vis Exp 2012:e2952. [PMID: 22664543 PMCID: PMC3466949 DOI: 10.3791/2952] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Human perivascular stem cells (PSCs) can be isolated in sufficient numbers from multiple tissues for purposes of skeletal tissue engineering1-3. PSCs are a FACS-sorted population of 'pericytes' (CD146+CD34-CD45-) and 'adventitial cells' (CD146-CD34+CD45-), each of which we have previously reported to have properties of mesenchymal stem cells. PSCs, like MSCs, are able to undergo osteogenic differentiation, as well as secrete pro-osteogenic cytokines1,2. In the present protocol, we demonstrate the osteogenicity of PSCs in several animal models including a muscle pouch implantation in SCID (severe combined immunodeficient) mice, a SCID mouse calvarial defect and a femoral segmental defect (FSD) in athymic rats. The thigh muscle pouch model is used to assess ectopic bone formation. Calvarial defects are centered on the parietal bone and are standardly 4 mm in diameter (critically sized)8. FSDs are bicortical and are stabilized with a polyethylene bar and K-wires4. The FSD described is also a critical size defect, which does not significantly heal on its own4. In contrast, if stem cells or growth factors are added to the defect site, significant bone regeneration can be appreciated. The overall goal of PSC xenografting is to demonstrate the osteogenic capability of this cell type in both ectopic and orthotopic bone regeneration models.
Collapse
Affiliation(s)
- Aaron W James
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, UCLA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Cowan CM, Zhang X, James AW, Kim TM, Sun N, Wu B, Ting K, Soo C. NELL-1 increases pre-osteoblast mineralization using both phosphate transporter Pit1 and Pit2. Biochem Biophys Res Commun 2012; 422:351-7. [PMID: 22580275 DOI: 10.1016/j.bbrc.2012.04.077] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 04/14/2012] [Indexed: 11/15/2022]
Abstract
NELL-1 is a potent osteoinductive molecule that enhances bone formation in multiple animal models through currently unidentified pathways. In the present manuscript, we hypothesized that NELL-1 may regulate osteogenic differentiation accompanied by alteration of inorganic phosphate (Pi) entry into the osteoblast via sodium dependent phosphate (NaPi) transporters. To determine this, MC3T3-E1 pre-osteoblasts were cultured in the presence of recombinant human (rh)NELL-1 or rhBMP-2. Analysis was performed for intracellular Pi levels through malachite green staining, Pit-1 and Pit-2 expression, and forced upregulation of Pit-1 and Pit-2. Results showed rhNELL-1 to increase MC3T3-E1 matrix mineralization and Pi influx associated with activation of both Pit-1 and Pit-2 channels, with significantly increased Pit-2 production. In contrast, Pi transport elicited by rhBMP-2 showed to be associated with increased Pit-1 production only. Next, neutralizing antibodies against Pit-1 and Pit-2 completely abrogated the Pi influx effect of rhNELL-1, suggesting rhNELL-1 is dependent on both transporters. These results identify one potential mechanism of action for rhNELL-1 induced osteogenesis and highlight a fundamental difference between NELL-1 and BMP-2 signaling.
Collapse
Affiliation(s)
- Catherine M Cowan
- Department of Bioengineering, University of California, Los Angeles, 420 Westwood Plaza,7523 Boelter Hall, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Hu J, Hou Y, Park H, Lee M. Beta-tricalcium phosphate particles as a controlled release carrier of osteogenic proteins for bone tissue engineering. J Biomed Mater Res A 2012; 100:1680-6. [PMID: 22447727 DOI: 10.1002/jbm.a.34115] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 10/14/2011] [Accepted: 02/01/2012] [Indexed: 11/12/2022]
Abstract
Beta-tricalcium phosphate (β-TCP) has been widely used as bone substitutes and delivery carriers of osteogenic proteins. However, low protein carrying capacity and agent burst release profiles of β-TCP limit their usage. This study investigates strategies to enhance protein carrying capacity of β-TCP particles with reduced initial burst by surface etching in citric acid solution or by creating apatite coatings with the simulate body fluid immersion approach. The release kinetics of protein from the modified β-TCP particles was investigated using Nel-like molecule-1 (Nell-1), a novel osteogenic protein, as a model protein. Although chemical etching treatments reduced the initial burst release of protein from the particles, a rapid burst release was observed with high protein dose. In contrast, the burst release of protein was significantly reduced by the apatite coating and a high protein dose was successfully delivered over a prolonged period from the apatite-coated particles. Protein release was further modulated by simultaneously delivering proteins from two different substrates: acid-etched and apatite-coated particles. The bioactivity of the protein was preserved during the loading procedure onto the particles. In addition, protein-loaded particles maintained biological activity in the lyophilized state over 4 weeks. These findings suggest that the protein carrying capacity of β-TCP can be modulated by surface modification, which has a potential for use as a protein carrier with controlled release.
Collapse
Affiliation(s)
- Junli Hu
- Division of Advanced Prosthodontics, Biomaterials and Hospital Dentistry, University of California, Los Angeles, California 90095, USA
| | | | | | | |
Collapse
|
47
|
James AW, Pang S, Askarinam A, Corselli M, Zara JN, Goyal R, Chang L, Pan A, Shen J, Yuan W, Stoker D, Zhang X, Adams JS, Ting K, Soo C. Additive effects of sonic hedgehog and Nell-1 signaling in osteogenic versus adipogenic differentiation of human adipose-derived stromal cells. Stem Cells Dev 2012; 21:2170-8. [PMID: 22264144 DOI: 10.1089/scd.2011.0461] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A theoretical inverse relationship exists between osteogenic (bone forming) and adipogenic (fat forming) mesenchymal stem cell (MSC) differentiation. This inverse relationship in theory partially underlies the clinical entity of osteoporosis, in which marrow MSCs have a preference for adipose differentiation that increases with age. Two pro-osteogenic cytokines have been recently studied that each also possesses antiadipogenic properties: Sonic Hedgehog (SHH) and NELL-1 proteins. In the present study, we assayed the potential additive effects of the biologically active N-terminus of SHH (SHH-N) and NELL-1 protein on osteogenic and adipogenic differentiation of human primary adipose-derived stromal cell (hASCs). We observed that both recombinant SHH-N and NELL-1 protein significantly enhanced osteogenic differentiation and reduced adipose differentiation across all markers examined (alkaline phosphatase, Alizarin red and Oil red O staining, and osteogenic gene expression). Moreover, SHH-N and NELL-1 directed signaling produced additive effects on the pro-osteogenic and antiadipogenic differentiation of hASCs. NELL-1 treatment increased Hedgehog signaling pathway expression; coapplication of the Smoothened antagonist Cyclopamine reversed the pro-osteogenic effect of NELL-1. In summary, Hedgehog and Nell-1 signaling exert additive effects on the pro-osteogenic and antiadipogenic differentiation of ASCs. These studies suggest that the combination cytokines SHH-N+NELL-1 may represent a viable future technique for inducing the osteogenic differentiation of MSCs.
Collapse
Affiliation(s)
- Aaron W James
- Section of Orthodontics, Dental and Craniofacial Research Institute, School of Dentistry, University of California, Los Angeles, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Siu RK, Zara JN, Hou Y, James AW, Kwak J, Zhang X, Ting K, Wu BM, Soo C, Lee M. NELL-1 promotes cartilage regeneration in an in vivo rabbit model. Tissue Eng Part A 2011; 18:252-61. [PMID: 21902605 DOI: 10.1089/ten.tea.2011.0142] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Repair of cartilage due to joint trauma remains challenging due to the poor healing capacity of cartilage and adverse effects related to current growth factor-based strategies. NELL-1 (Nel-like molecule-1; Nel [a protein strongly expressed in neural tissue encoding epidermal growth factor like domain]), a protein first characterized in the context of premature cranial suture fusion, is believed to accelerate differentiation along the osteochondral lineage. We previously demonstrated the ability of NELL-1 protein to maintain the cartilaginous phenotype of explanted rabbit chondrocytes in vitro. Our objective in the current study is to determine whether NELL-1 can affect endogenous chondrocytes in an in vivo cartilage defect model. To generate the implant, NELL-1 was incorporated into chitosan nanoparticles and embedded into alginate hydrogels. These implants were press fit into 3-mm circular osteochondral defects created in the femoral condylar cartilage of 3-month-old New Zealand White rabbits (n=10). Controls included unfilled defects (n=8) and defects filled with phosphate-buffered saline-loaded chitosan nanoparticles embedded in alginate hydrogels (n=8). Rabbits were sacrificed 3 months postimplantation for histological analysis. Defects filled with alginate containing NELL-1 demonstrated significantly improved cartilage regeneration. Remarkably, histology of NELL-1-treated defects closely resembled that of native cartilage, including stronger Alcian blue and Safranin-O staining and increased deposition of type II collagen and absence of the bone markers type I collagen and Runt-related transcription factor 2 (Runx2) as demonstrated by immunohistochemistry. Our results suggest that NELL-1 may produce functional cartilage with properties similar to native cartilage, and is an exciting candidate for tissue engineering-based approaches for treating diverse pathologies of cartilage defects and degeneration.
Collapse
Affiliation(s)
- Ronald K Siu
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
James AW, Pan A, Chiang M, Zara J, Zhang X, Ting K, Soo C. A new function of Nell-1 protein in repressing adipogenic differentiation. Biochem Biophys Res Commun 2011; 411:126-31. [PMID: 21723263 PMCID: PMC3166249 DOI: 10.1016/j.bbrc.2011.06.111] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 06/15/2011] [Indexed: 12/25/2022]
Abstract
A theoretical inverse relationship has long been postulated for osteogenic and adipogenic differentiation (bone versus adipose tissue differentiation). This inverse relationship in theory at least partially underlies the clinical entity of osteoporosis, in which marrow mesenchymal stem cells (MSCs) have a predilection for adipose differentiation that increases with age. In the present study, we assayed the potential anti-adipogenic effects of Nell-1 protein (an osteoinductive molecule). Using 3T3-L1 (a human preadipocyte cell line) cells and human adipose-derived stromal cells (ASCs), we observed that adenoviral delivered (Ad)-Nell-1 or recombinant NELL-1 protein significantly reduced adipose differentiation across all markers examined (Oil red O staining, adipogenic gene expression [Pparg, Lpl, Ap2]). In a prospective fashion, Hedgehog signaling was assayed as potentially downstream of Nell-1 signaling in regulating osteogenic over adipogenic differentiation. In comparison to Ad-LacZ control, Ad-Nell-1 increased expression of hedgehog signaling markers (Ihh, Gli1, Ptc1). These studies suggest that Nell-1 is a potent anti-adipogenic agent. Moreover, Nell-1 signaling may inhibit adipogenic differentiation via a Hedgehog dependent mechanism.
Collapse
Affiliation(s)
- Aaron W. James
- Dental and Craniofacial Research Institute, University of California, Los Angeles, Los Angeles CA 90095
- Section of Orthodontics School of Dentistry, University of California, Los Angeles, Los Angeles CA 90095
- Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles CA 90095
| | - Angel Pan
- Dental and Craniofacial Research Institute, University of California, Los Angeles, Los Angeles CA 90095
| | - Michael Chiang
- Dental and Craniofacial Research Institute, University of California, Los Angeles, Los Angeles CA 90095
- Section of Orthodontics School of Dentistry, University of California, Los Angeles, Los Angeles CA 90095
| | - Janette Zara
- Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles CA 90095
| | - Xinli Zhang
- Dental and Craniofacial Research Institute, University of California, Los Angeles, Los Angeles CA 90095
- Section of Orthodontics School of Dentistry, University of California, Los Angeles, Los Angeles CA 90095
| | - Kang Ting
- Dental and Craniofacial Research Institute, University of California, Los Angeles, Los Angeles CA 90095
- Section of Orthodontics School of Dentistry, University of California, Los Angeles, Los Angeles CA 90095
- Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles CA 90095
| | - Chia Soo
- Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles CA 90095
| |
Collapse
|