1
|
Tuersuntuoheti M, Zhang J, Zhou W, Zhang CL, Liu C, Chang Q, Liu S. Exploring the growth trait molecular markers in two sheep breeds based on Genome-wide association analysis. PLoS One 2023; 18:e0283383. [PMID: 36952432 PMCID: PMC10035858 DOI: 10.1371/journal.pone.0283383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 03/08/2023] [Indexed: 03/25/2023] Open
Abstract
Growth traits are quantitative traits controlled by multiple micro-effect genes. we identified molecular markers related to sheep growth traits, which formed the basis of molecular breeding. In this study, we randomly selected 100 Qira Black sheep and 84 German Merino sheep for the blood collection the jugular vein to genotype by using the Illumina Ovine SNP 50K Bead Chip. quality control criteria for statistical analysis were: rejection detection rate < 90% and minimum allele frequency (MAF) < 5%. Then, we performed Genome-wide association studies (GWAS) on sheep body weight, body height, body length, and chest circumference using mixed linear models. After getting 55 SNPs with significant correlation, they were annotated by reference genome of Ovis aries genome (Oar_v4.0) and We obtained a total of 84 candidate genes associated with production traits (BMPR1B, HSD17B3, TMEM63C, etc.). We selected BMPR1B for population validation and found a correlation between the FecB locus and body weight traits. Therefore, this study not only supplements the existing knowledge of molecular markers of sheep growth traits, but also has important theoretical significance and reference value for the mining of functional genes of sheep growth traits.
Collapse
Affiliation(s)
- Mirenisa Tuersuntuoheti
- College of Animal Science and Technology, Tarim University, Alar, China
- Tarim Science and Technology Key Laboratory of Xinjiang Production and Construction Corps, Alar, China
| | - Jihu Zhang
- College of Animal Science and Technology, Tarim University, Alar, China
- Tarim Science and Technology Key Laboratory of Xinjiang Production and Construction Corps, Alar, China
| | - Wen Zhou
- College of Animal Science and Technology, Tarim University, Alar, China
- Tarim Science and Technology Key Laboratory of Xinjiang Production and Construction Corps, Alar, China
| | - Cheng-Long Zhang
- College of Animal Science and Technology, Tarim University, Alar, China
- Tarim Science and Technology Key Laboratory of Xinjiang Production and Construction Corps, Alar, China
| | - Chunjie Liu
- College of Animal Science and Technology, Tarim University, Alar, China
- Tarim Science and Technology Key Laboratory of Xinjiang Production and Construction Corps, Alar, China
| | - Qianqian Chang
- College of Animal Science and Technology, Tarim University, Alar, China
- Tarim Science and Technology Key Laboratory of Xinjiang Production and Construction Corps, Alar, China
| | - Shudong Liu
- College of Animal Science and Technology, Tarim University, Alar, China
- Tarim Science and Technology Key Laboratory of Xinjiang Production and Construction Corps, Alar, China
| |
Collapse
|
2
|
Zeng L, He H, Sun M, Gong X, Zhou M, Hong Y, Wu Y, Chen X, Chen Q. Runx2 and Nell-1 in dental follicle progenitor cells regulate bone remodeling and tooth eruption. Stem Cell Res Ther 2022; 13:486. [PMID: 36175952 PMCID: PMC9524038 DOI: 10.1186/s13287-022-03140-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/15/2022] [Indexed: 11/10/2022] Open
Abstract
Dental follicles are necessary for tooth eruption, surround the enamel organ and dental papilla, and regulate both the formation and resorption of alveolar bone. Dental follicle progenitor cells (DFPCs), which are stem cells found in dental follicles, differentiate into different kinds of cells that are necessary for tooth formation and eruption. Runt‐related transcription factor 2 (Runx2) is a transcription factor that is essential for osteoblasts and osteoclasts differentiation, as well as bone remodeling. Mutation of Runx2 causing cleidocranial dysplasia negatively affects osteogenesis and the osteoclastic ability of dental follicles, resulting in tooth eruption difficulties. Among a variety of cells and molecules, Nel-like molecule type 1 (Nell-1) plays an important role in neural crest-derived tissues and is strongly expressed in dental follicles. Nell-1 was originally identified in pathologically fused and fusing sutures of patients with unilateral coronal synostosis, and it plays indispensable roles in bone remodeling, including roles in osteoblast differentiation, bone formation and regeneration, craniofacial skeleton development, and the differentiation of many kinds of stem cells. Runx2 was proven to directly target the Nell-1 gene and regulate its expression. These studies suggested that Runx2/Nell-1 axis may play an important role in the process of tooth eruption by affecting DFPCs. Studies on short and long regulatory noncoding RNAs have revealed the complexity of RNA-mediated regulation of gene expression at the posttranscriptional level. This ceRNA network participates in the regulation of Runx2 and Nell-1 gene expression in a complex way. However, non-study indicated the potential connection between Runx2 and Nell-1, and further researches are still needed.
Collapse
Affiliation(s)
- Li Zeng
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Hong He
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China. .,Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China.
| | - Mingjie Sun
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Xinyi Gong
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Mengqi Zhou
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Yaya Hong
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Yongjia Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Xuepeng Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China. .,Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China.
| | - Qianming Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China. .,Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China.
| |
Collapse
|
3
|
Cheng X, Shi J, Jia Z, Ha P, Soo C, Ting K, James AW, Shi B, Zhang X. NELL-1 in Genome-Wide Association Studies across Human Diseases. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:395-405. [PMID: 34890556 PMCID: PMC8895422 DOI: 10.1016/j.ajpath.2021.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 11/19/2021] [Accepted: 11/24/2021] [Indexed: 02/08/2023]
Abstract
Neural epidermal growth factor-like (EGFL)-like protein (NELL)-1 is a potent and key osteogenic factor in the development and regeneration of skeletal tissues. Intriguingly, accumulative data from genome-wide association studies (GWASs) have started unveiling potential broader roles of NELL-1 beyond its functions in bone and cartilage. With exploration of the genetic variants of the entire genome in large-scale disease cohorts, GWASs have been used for establishing the connection between specific single-nucleotide polymorphisms of NELL1, in addition to osteoporosis, metabolic diseases, inflammatory conditions, neuropsychiatric diseases, neurodegenerative disorders, and malignant tumors. This review summarizes the findings from GWASs on the manifestation, significance level, implications on function, and correlation of specific NELL1 single-nucleotide polymorphisms in various disorders in humans. By offering a unique and comprehensive correlation between genetic variants and plausible functions of NELL1 in GWASs, this review illustrates the wide range of potential effects of a single gene on the pathogenesis of multiple disorders in humans.
Collapse
Affiliation(s)
- Xu Cheng
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, and the Department of Cleft Lip and Palate, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California-Los Angeles, Los Angeles, California
| | - Jiayu Shi
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California-Los Angeles, Los Angeles, California
| | - Zhonglin Jia
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, and the Department of Cleft Lip and Palate, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Pin Ha
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California-Los Angeles, Los Angeles, California
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, Orthopaedic Hospital Research Center, University of California-Los Angeles, Los Angeles, California
| | - Kang Ting
- Forsyth Institute, affiliate of the Harvard School of Dental Medicine, Boston, Massachusetts
| | - Aaron W James
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Bing Shi
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, and the Department of Cleft Lip and Palate, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Xinli Zhang
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California-Los Angeles, Los Angeles, California.
| |
Collapse
|
4
|
Kague E, Medina-Gomez C, Boyadjiev SA, Rivadeneira F. The genetic overlap between osteoporosis and craniosynostosis. Front Endocrinol (Lausanne) 2022; 13:1020821. [PMID: 36225206 PMCID: PMC9548872 DOI: 10.3389/fendo.2022.1020821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/08/2022] [Indexed: 11/29/2022] Open
Abstract
Osteoporosis is the most prevalent bone condition in the ageing population. This systemic disease is characterized by microarchitectural deterioration of bone, leading to increased fracture risk. In the past 15 years, genome-wide association studies (GWAS), have pinpointed hundreds of loci associated with bone mineral density (BMD), helping elucidate the underlying molecular mechanisms and genetic architecture of fracture risk. However, the challenge remains in pinpointing causative genes driving GWAS signals as a pivotal step to drawing the translational therapeutic roadmap. Recently, a skull BMD-GWAS uncovered an intriguing intersection with craniosynostosis, a congenital anomaly due to premature suture fusion in the skull. Here, we recapitulate the genetic contribution to both osteoporosis and craniosynostosis, describing the biological underpinnings of this overlap and using zebrafish models to leverage the functional investigation of genes associated with skull development and systemic skeletal homeostasis.
Collapse
Affiliation(s)
- Erika Kague
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, Bristol, United Kingdom
- *Correspondence: Erika Kague,
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus Medical Center (MC), University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Simeon A. Boyadjiev
- Department of Pediatrics, University of California, Davis, Sacramento, CA, United States
| | - Fernando Rivadeneira
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Center (MC), University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
5
|
Gao C, Liu H, Zhao Y, Miao X, Zheng H. Is there a relationship between neural EGFL like 1 (NELL1) promoter hypermethylation and prognosis of gastric cancer? Med Hypotheses 2021; 158:110723. [PMID: 34753006 DOI: 10.1016/j.mehy.2021.110723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/14/2021] [Accepted: 10/07/2021] [Indexed: 11/19/2022]
Abstract
We hypothesized that neural EGFL like 1 (NELL1) promoter hypermethylation might be associated with the prognosis of gastric cancer. Some studies considered NELL1 as a tumor suppressor gene and our research confirmed for the first time the hypermethylation in the promoter region of NELL1 by the application of mass spectrometry. Promoter hypermethylation can cause the silencing of tumor suppressor genes and promote tumor progression. Based on present studies and research results, we proposed that NELL1 promoter hypermethylation might be associated with cancer staging and the survival of gastric cancer patients and had prognostic value. We hoped that NELL1 promoter hypermethylation would be applied not only for early detection but also prognosis prediction of gastric cancer and would become a new prognostic biomarker.
Collapse
Affiliation(s)
- Changlu Gao
- The Fourth Affiliated Hospital of Harbin Medical University, No. 37, Yiyuan Street, Nangang District, Harbin, China
| | - Haibin Liu
- The Fourth Affiliated Hospital of Harbin Medical University, No. 37, Yiyuan Street, Nangang District, Harbin, China
| | - Yubo Zhao
- The Fourth Affiliated Hospital of Harbin Medical University, No. 37, Yiyuan Street, Nangang District, Harbin, China
| | - Xinyu Miao
- The Fourth Affiliated Hospital of Harbin Medical University, No. 37, Yiyuan Street, Nangang District, Harbin, China
| | - Hongqun Zheng
- The Fourth Affiliated Hospital of Harbin Medical University, No. 37, Yiyuan Street, Nangang District, Harbin, China.
| |
Collapse
|
6
|
Mechanisms of Primary Membranous Nephropathy. Biomolecules 2021; 11:biom11040513. [PMID: 33808418 PMCID: PMC8065962 DOI: 10.3390/biom11040513] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 12/11/2022] Open
Abstract
Membranous nephropathy (MN) is an autoimmune disease of the kidney glomerulus and one of the leading causes of nephrotic syndrome. The disease exhibits heterogenous outcomes with approximately 30% of cases progressing to end-stage renal disease. The clinical management of MN has steadily advanced owing to the identification of autoantibodies to the phospholipase A2 receptor (PLA2R) in 2009 and thrombospondin domain-containing 7A (THSD7A) in 2014 on the podocyte surface. Approximately 50–80% and 3–5% of primary MN (PMN) cases are associated with either anti-PLA2R or anti-THSD7A antibodies, respectively. The presence of these autoantibodies is used for MN diagnosis; antibody levels correlate with disease severity and possess significant biomarker values in monitoring disease progression and treatment response. Importantly, both autoantibodies are causative to MN. Additionally, evidence is emerging that NELL-1 is associated with 5–10% of PMN cases that are PLA2R- and THSD7A-negative, which moves us one step closer to mapping out the full spectrum of PMN antigens. Recent developments suggest exostosin 1 (EXT1), EXT2, NELL-1, and contactin 1 (CNTN1) are associated with MN. Genetic factors and other mechanisms are in place to regulate these factors and may contribute to MN pathogenesis. This review will discuss recent developments over the past 5 years.
Collapse
|
7
|
Mehta AS, Ha P, Zhu K, Li S, Ting K, Soo C, Zhang X, Zhao M. Physiological electric fields induce directional migration of mammalian cranial neural crest cells. Dev Biol 2021; 471:97-105. [PMID: 33340512 PMCID: PMC7856271 DOI: 10.1016/j.ydbio.2020.12.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/23/2022]
Abstract
During neurulation, cranial neural crest cells (CNCCs) migrate long distances from the neural tube to their terminal site of differentiation. The pathway traveled by the CNCCs defines the blueprint for craniofacial construction, abnormalities of which contribute to three-quarters of human birth defects. Biophysical cues like naturally occurring electric fields (EFs) have been proposed to be one of the guiding mechanisms for CNCC migration from the neural tube to identified position in the branchial arches. Such endogenous EFs can be mimicked by applied EFs of physiological strength that has been reported to guide the migration of amphibian and avian neural crest cells (NCCs), namely galvanotaxis or electrotaxis. However, the behavior of mammalian NCCs in external EFs has not been reported. We show here that mammalian CNCCs migrate towards the anode in direct current (dc) EFs. Reversal of the field polarity reverses the directedness. The response threshold was below 30 mV/mm and the migration directedness and displacement speed increased with increase in field strength. Both CNCC line (O9-1) and primary mouse CNCCs show similar galvanotaxis behavior. Our results demonstrate for the first time that the mammalian CNCCs respond to physiological EFs by robust directional migration towards the anode in a voltage-dependent manner.
Collapse
Affiliation(s)
- Abijeet Singh Mehta
- Department of Ophthalmology & Vision Science, Institute for Regenerative Cures, Center for Neuroscience, University of California at Davis, School of Medicine, Suite 1630, Room 1617, 2921 Stockton Blvd., Sacramento, CA, 95817, USA; Department of Dermatology, University of California, Davis, CA, USA
| | - Pin Ha
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Kan Zhu
- Department of Ophthalmology & Vision Science, Institute for Regenerative Cures, Center for Neuroscience, University of California at Davis, School of Medicine, Suite 1630, Room 1617, 2921 Stockton Blvd., Sacramento, CA, 95817, USA; Department of Dermatology, University of California, Davis, CA, USA
| | - ShiYu Li
- Department of Ophthalmology & Vision Science, Institute for Regenerative Cures, Center for Neuroscience, University of California at Davis, School of Medicine, Suite 1630, Room 1617, 2921 Stockton Blvd., Sacramento, CA, 95817, USA; Department of Dermatology, University of California, Davis, CA, USA
| | - Kang Ting
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, 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, CA, 90095, USA
| | - Xinli Zhang
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, CA, USA.
| | - Min Zhao
- Department of Ophthalmology & Vision Science, Institute for Regenerative Cures, Center for Neuroscience, University of California at Davis, School of Medicine, Suite 1630, Room 1617, 2921 Stockton Blvd., Sacramento, CA, 95817, USA; Department of Dermatology, University of California, Davis, CA, USA.
| |
Collapse
|
8
|
Elarjani T, Almutairi OT, Alhussinan M, Alturkistani A, Alotaibi FS, Bafaquh M, Alotaibi FE. Bibliometric analysis of the top 100 most cited articles on craniosynostosis. Childs Nerv Syst 2021; 37:587-597. [PMID: 32780272 DOI: 10.1007/s00381-020-04858-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/06/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Craniosynostosis is the premature closure of cranial sutures and it continues to be a therapeutic challenge due to the diversity and complexity of the syndrome. Bibliometric analysis is a study of ranking citations and exploring the most impactful articles in a respective discipline. It also demonstrates the chronological trends of publications. METHODS In May 2020, we performed a title-specific search of the Scopus database using "craniosynostosis" as our query term without publication date restrictions. The top 100 articles in craniosynostosis were retrieved and analyzed. RESULTS The top 100 most-cited articles in craniosynostosis received a total 13,826 citations, and an average of 138 citations per paper. The publication dates ranged from 1920 to 2015, with a peak period of top publications between 1996 and 2005. The most common category is clinical, followed by neurogenetics. The top cited article received 540 citation counts and 19.29 citations per year. The USA was the most contributing country to the list. The Journal of Plastic and Reconstructive Surgery published the largest number of top cited articles. Neurosurgery as a specialty contributed to most articles in the list (27 articles). The institute who contributed the most was the Assistance Publique Hopitaux Paris. CONCLUSION Bibliometric analysis in craniosynostosis revealed major trend changes of research over the years, with a focus on neurogenetics and the different types of surgical corrections. The current collection of highly cited publications may assist physicians in gaining a better understanding of the evidence-based approach in craniosynostosis.
Collapse
Affiliation(s)
- Turki Elarjani
- Department of Neurological Surgery, University of Miami, Miami, FL, USA.
| | - Othman T Almutairi
- Division of Neurological Surgery, Neurosciences Department, King Fahad Medical City, Riyadh, Saudi Arabia
| | | | - Abdulelah Alturkistani
- Division of Neurological Surgery, Neurosciences Department, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Fahad S Alotaibi
- Division of Neurological Surgery, Neurosciences Department, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Mohammed Bafaquh
- Division of Neurological Surgery, Neurosciences Department, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Fahad E Alotaibi
- Division of Neurological Surgery, Neurosciences Department, King Fahad Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
9
|
Xu C, Xu J, Zhang X, Xu S, Liu Q, Weng Z, Gu A. Serum nickel is associated with craniosynostosis risk: Evidence from humans and mice. ENVIRONMENT INTERNATIONAL 2021; 146:106289. [PMID: 33276314 DOI: 10.1016/j.envint.2020.106289] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/15/2020] [Accepted: 11/16/2020] [Indexed: 05/06/2023]
Abstract
BACKGROUND To date, few studies have explored the effects of exposure to metal mixtures on adverse developmental outcomes, and no reported studies have linked metal exposure to craniosynostosis (CS). The purpose of this study is to investigate the association between metal exposure and the risk of CS by conducting epidemiological and experimental studies. METHODS Inductively coupled plasma mass spectrometry (ICP-MS) was used to measure the concentrations of 6 metals (chromium [Cr], nickel [Ni], tin [Sn], arsenic [As], thallium [Tl], and lead [Pb]) in serum samples from 174 CS patients and 85 control individuals. Non-syndromic patients with isolated sagittal suture closure were selected as the case group, and healthy children matched by sex and age were selected as controls. Bayesian kernel machine regression (BKMR) models were used to account for joint metal effects. Multiple logistic regression analysis was used to explore the association between metal concentration and CS occurrence, with adjustment for potential confounders. During pregnancy, mice were exposed to Ni (0, 0.05, or 0.1 g/kg/day) until weaning, and the widths of the sutures and shapes of the skull were analysed by micro-CT 3D imaging and histological analysis. MC3T3-E1 cells were treated with Ni (0, 0.005, or 0.05 μg/mL) for 72 h. Alkaline phosphate (ALP) staining and Alizarin red staining were performed to observe the development of osteoblasts. The expression levels of osteoblast-related genes were also detected. RESULTS A positive association between the metal mixture and CS risk was observed based on population data; the Ni group had the highest conditional posterior inclusion probability (PIP), at 0.8416, and in the fully adjusted model, the highest Ni exposure level had a more significant association with CS (coefficient = 2.65, 95% CI: 0.29, 5.02) than the lowest Ni exposure level. The mean widths of the sagittal sutures in mice were 8.8 ± 0.6 mm in the control group, 8.0 ± 0.8 mm in the 0.05 g/kg/day group and 6.8 ± 0.4 mm in the 0.1 g/kg/day group. After Ni exposure, ALP gene expression in skull tissue was increased, and ALP activity was increased in MC3T3-E1 cells. Moreover, increased collagen content in mouse skull sections and elevated osteocalcin (OCN) expression in MC3T3-E1 cells were observed in the Ni-treated groups compared to the control group. CONCLUSIONS This study is the first to provide evidence that increased serum Ni was associated with an increased risk of CS. Early life exposure to Ni promoted osteogenesis during skull growth, which may contribute to the development of CS.
Collapse
Affiliation(s)
- Cheng Xu
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of the Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Jin Xu
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of the Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, China; Department of Maternal, Child, and Adolescent Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xin Zhang
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of the Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Shuqin Xu
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of the Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Qian Liu
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of the Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Zhenkun Weng
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of the Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Aihua Gu
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of the Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
10
|
Lai K, Xi Y, Du X, Jiang Z, Li Y, Huang T, Miao X, Wang H, Wang Y, Yang G. Activation of Nell-1 in BMSC Sheet Promotes Implant Osseointegration Through Regulating Runx2/Osterix Axis. Front Cell Dev Biol 2020; 8:868. [PMID: 33072736 PMCID: PMC7536315 DOI: 10.3389/fcell.2020.00868] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/11/2020] [Indexed: 12/11/2022] Open
Abstract
Neural epidermal growth factor-like 1 protein (Nell-1) is first studied because of its association with human craniosynostosis. Nell-1 has been used to accelerate the process of fracture healing because of the osteoinductive ability in recent years. However, the role of Nell-1 during the process of osteointegration is unknown. Here we show that activation of Nell-1 in the BMSC sheet promotes osseointegration in vivo and in vitro. We found that overexpression of Nell-1 improved osteogenic differentiation and enhanced matrix mineralization of BMSCs through increasing expression of Runx2 and Osterix. Activation of Nell-1 up-regulated the expression ratio of OPG/RANKL, which might have a negative influence on osteoclast differentiation. Furthermore, we obtained BMSC sheet-implant complexes transfected with lentivirus overexpressing and interfering Nell-1 in in vivo study, and confirmed that overexpression of Nell-1 promoted new bone formation around the implant and increased the bone-implant contacting area percentage. Our results demonstrate that activation of Nell-1 improves implant osteointegration by regulating Runx2/Osterix axis and shows the potential of BMSC sheet-implant complexes in gene therapy.
Collapse
Affiliation(s)
- Kaichen Lai
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Yue Xi
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Xue Du
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Zhiwei Jiang
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Yongzheng Li
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Tingben Huang
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Xiaoyan Miao
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Huiming Wang
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Ying Wang
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Guoli Yang
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| |
Collapse
|
11
|
Liu Y, Ju M, Wang Z, Li J, Shao C, Fu T, Jing Y, Zhao Y, Lv Z, Li G. The synergistic effect of NELL1 and adipose-derived stem cells on promoting bone formation in osteogenesis imperfecta treatment. Biomed Pharmacother 2020; 128:110235. [PMID: 32454289 DOI: 10.1016/j.biopha.2020.110235] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/30/2020] [Accepted: 05/03/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Osteogenesis imperfecta (OI) is a rare genetic disorder characterized by bone fragility and deformity. Mesenchymal stem cells (MSCs) infusion can improve bone performance mainly due to their differentiation into osteoblasts in OI therapy. The osteoinductive activity of NELL1 have benefited various bone defect and osteoporotic models by promoting bone formation. The present study investigated the efficacy of combined use of NELL1 and adipose-derived mesenchymal stem cells (ADSCs) in OI treatment. METHODS Lentiviral vector carrying mouse Nell1 gene was constructed and lentivirus were used to infect ADSCs. The osteogenic capacity of MC3T3-E1 and ADSCs stimulated by recombinant mouse NELL1 protein (rmNELL1) and Nell1 gene genetically modified ADSCs (lenti-Nell1-ADSCs) were estimated by real-time quantitative PCR. Thirty adult male OI type I mice with single Col1a1 gene knockout were randomly divided into five groups and received intravenously injected PBS, rmNELL1 (1.25 mg/Kg), ADSCs (2 × 105 cells per mice), rmNELL1 (1.25 mg/Kg) combined with ADSCs (2 × 105 cells per mice), or lenti-Nell1-ADSCs (2 × 105 cells per mice) respectively. Six wildtype (WT) mice served as positive control. Bone formation was examined after 4 weeks using micro-CT, histological and immunohistochemical methods. RESULTS Three osteoblast related genes of MC3T3-E1 and ADSCs were significantly up-regulated by rmNELL1 in vitro. Lenti-Nell1-ADSCs showed greatly enhanced osteogenic differentiation capacity. The infused lenti-Nell1-ADSCs could migrate to femur and differentiate into ALPL-positive cells. Systemic administration of rmNELL1 combined with ADSCs or lenti-Nell1-ADSCs markedly improved the femoral microstructure and promoted bone formation through increasing the ALPL and osteocalcin (OCN) expression, much better than mice that received single rmNELL1 or ADSCs. And Nell1 gene engineered ADSCs achieved slightly better outcomes than that of combinative use of rmNELL1 and ADSCs. CONCLUSIONS NELL1 and ADSCs exhibited synergistic effect on stimulating bone formation of OI mice, which might provide an alternative strategy in OI treatment. Compared with dose escalation or multiple administration of rmNELL1, lentivirus-mediated long term expression of NELL1 might be more feasible and convenient. However, further studies are needed to confirm the safety and optimize the therapeutic regime.
Collapse
Affiliation(s)
- Yi Liu
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Mingyan Ju
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Zihan Wang
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Jiaci Li
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Chenyi Shao
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Ting Fu
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Yaqing Jing
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Yuxia Zhao
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Zhe Lv
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Guang Li
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, People's Republic of China.
| |
Collapse
|
12
|
Chen X, Wang H, Yu M, Kim JK, Qi H, Ha P, Jiang W, Chen E, Luo X, Needle RB, Baik L, Yang C, Shi J, Kwak JH, Ting K, Zhang X, Soo C. Cumulative inactivation of Nell-1 in Wnt1 expressing cell lineages results in craniofacial skeletal hypoplasia and postnatal hydrocephalus. Cell Death Differ 2020; 27:1415-1430. [PMID: 31582804 PMCID: PMC7206096 DOI: 10.1038/s41418-019-0427-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 07/09/2019] [Accepted: 08/26/2019] [Indexed: 02/05/2023] Open
Abstract
Upregulation of Nell-1 has been associated with craniosynostosis (CS) in humans, and validated in a mouse transgenic Nell-1 overexpression model. Global Nell-1 inactivation in mice by N-ethyl-N-nitrosourea (ENU) mutagenesis results in neonatal lethality with skeletal abnormalities including cleidocranial dysplasia (CCD)-like calvarial bone defects. This study further defines the role of Nell-1 in craniofacial skeletogenesis by investigating specific inactivation of Nell-1 in Wnt1 expressing cell lineages due to the importance of cranial neural crest cells (CNCCs) in craniofacial tissue development. Nell-1flox/flox; Wnt1-Cre (Nell-1Wnt1 KO) mice were generated for comprehensive analysis, while the relevant reporter mice were created for CNCC lineage tracing. Nell-1Wnt1 KO mice were born alive, but revealed significant frontonasal and mandibular bone defects with complete penetrance. Immunostaining demonstrated that the affected craniofacial bones exhibited decreased osteogenic and Wnt/β-catenin markers (Osteocalcin and active-β-catenin). Nell-1-deficient CNCCs demonstrated a significant reduction in cell proliferation and osteogenic differentiation. Active-β-catenin levels were significantly low in Nell-1-deficient CNCCs, but were rescued along with osteogenic capacity to a level close to that of wild-type (WT) cells via exogenous Nell-1 protein. Surprisingly, 5.4% of young adult Nell-1Wnt1 KO mice developed hydrocephalus with premature ossification of the intrasphenoidal synchondrosis and widened frontal, sagittal, and coronal sutures. Furthermore, the epithelial cells of the choroid plexus and ependymal cells exhibited degenerative changes with misplaced expression of their respective markers, transthyretin and vimentin, as well as dysregulated Pit-2 expression in hydrocephalic Nell-1Wnt1 KO mice. Nell-1Wnt1 KO embryos at E9.5, 14.5, 17.5, and newborn mice did not exhibit hydrocephalic phenotypes grossly and/or histologically. Collectively, Nell-1 is a pivotal modulator of CNCCs that is essential for normal development and growth of the cranial vault and base, and mandibles partially via activating the Wnt/β-catenin pathway. Nell-1 may also be critically involved in regulating cerebrospinal fluid homeostasis and in the pathogenesis of postnatal hydrocephalus.
Collapse
Affiliation(s)
- Xiaoyan Chen
- Department of Orthodontics, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, PR China
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Huiming Wang
- Department of Oral Implantology, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Mengliu Yu
- Department of Oral Implantology, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, PR China
- Center of Stomatology, China-Japan Friendship Hospital, 2nd Yinghuayuan East Street, Chaoyang District, Beijing, PR China
| | - Jong Kil Kim
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Huichuan Qi
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, Changchun, Jilin, PR China
| | - Pin Ha
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Wenlu Jiang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Eric Chen
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Xiangyou Luo
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
- Department of Cleft Lip and Palate Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, PR China
| | - Ryan Brent Needle
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Lloyd Baik
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Cathryn Yang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Jiejun Shi
- Department of Orthodontics, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Jin Hee Kwak
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Kang Ting
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Xinli Zhang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA.
| | - Chia Soo
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA and Orthopaedic Hospital, University of California, Los Angeles, CA, USA
- UCLA Division of Plastic and Reconstructive Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, CA, USA
| |
Collapse
|
13
|
Yu L, Cen X, Xia K, Huang X, Sun W, Zhao Z, Liu J. microRNA expression profiles and the potential competing endogenous RNA networks in NELL-1-induced human adipose-derived stem cell osteogenic differentiation. J Cell Biochem 2020; 121:4623-4641. [PMID: 32065449 DOI: 10.1002/jcb.29695] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 01/27/2020] [Indexed: 02/05/2023]
Abstract
Studies have indicated that Nel-like molecule-1 (NELL-1) was an osteoblast-specific cytokine and some specific microRNAs (miRNAs) could serve as competing endogenous RNA (ceRNA) to partake in osteogenic differentiation of human adipose-derived stem cells (hASCs). The aim of this study was to explore the potential functional mechanisms of recombinant human NELL-1 protein (rhNELL-1) during hASCs osteogenic differentiation. rhNELL-1 was added to osteogenic medium to activate osteogenic differentiation of hASCs. High-throughput RNA sequencing (RNA-Seq) was performed and validated by real-time quantitative polymerase chain reaction. Gene ontology functional annotation and Kyoto Encyclopedia of Genes and Genomes pathway analysis were performed to detect the functions of differentially expressed miRNAs and genes. Coding-noncoding gene co-expression network and ceRNA networks were constructed to predict the potential regulatory role of miRNAs. A total of 1010 differentially expressed miRNAs and 1762 differentially expressed messenger RNAs (mRNAs) were detected. miRNA-370-3p, bone morphogenetic protein 2 (BMP2), and parathyroid hormone like hormone (PTHLH) were differentially expressed during NELL-1-induced osteogenesis. Bioinformatic analyses demonstrated that these differentially expressed miRNAs and mRNAs enriched in Rap1 signaling pathway, PI3K-Akt signaling pathway, p53 signaling pathway, Glucagon signaling pathway, and hypoxia-inducible factor-1 signaling pathway, which were important pathways related to osteogenic differentiation. In addition, miRNA-370-3p and has-miR-485-5p were predicted to interact with circ0001543, circ0002405, and ENST00000570267 in ceRNA networks. Based on the gain or loss of functional experiments by transfection, the results showed that miR-370-3p was a key regulator in osteogenic differentiation by targeting BMP2 and disturbing the expression of PTHLH, and participated in NELL-1-stimulated osteogenesis. The present study provided the primary data and evidence for further exploration on the roles of miRNAs and ceRNAs during NELL-1-induced ossification of hASCs.
Collapse
Affiliation(s)
- Liyuan Yu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xiao Cen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Temporomandibular Joint, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Kai Xia
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xinqi Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Wentian Sun
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jun Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
14
|
Niimi T. Roles of Slit Ligands and Their Roundabout (Robo) Family of Receptors in Bone Remodeling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 21:143-154. [PMID: 32986130 DOI: 10.1007/5584_2020_586] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Slit guidance ligands (Slits) and their roundabout (Robo) family of receptors are well-known axon guidance molecules that were originally identified in Drosophila mutants with commissural axon pathfinding defects. However, Slit-Robo signaling has been shown to be involved in not only neurogenesis, but also the development of other organs such as the kidney and heart. Recently, it was also revealed that Slit-Robo signaling plays an important role in bone metabolism. For example, osteoclast-derived Slit3 plays an osteoprotective role by synchronously stimulating bone formation by osteoblasts and suppressing bone resorption by osteoclasts through Robo receptors expressed on osteoblastic and osteoclastic cell lineages, making it a potential therapeutic target for metabolic bone disorders. Furthermore, osteoblast-derived Slit3 promotes bone formation indirectly as a proangiogenic factor. This review summarizes the recent progress on defining the roles of the Slit-Robo signaling in bone metabolism, and discusses the possible roles of the interaction between Robo and neural epidermal growth factor-like (NEL)-like (NELL) proteins that are novel ligands for Robo receptors.
Collapse
Affiliation(s)
- Tomoaki Niimi
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan.
| |
Collapse
|
15
|
Huang X, Cen X, Zhang B, Liao Y, Zhao Z, Zhu G, Zhao Z, Liu J. The roles of circRFWD2 and circINO80 during NELL-1-induced osteogenesis. J Cell Mol Med 2019; 23:8432-8441. [PMID: 31633307 PMCID: PMC6850935 DOI: 10.1111/jcmm.14726] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 08/27/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
Bone defects caused heavy social and economic burdens worldwide. Nel‐like molecule, type 1 (NELL‐1) could enhance the osteogenesis and the repairment of bone defects, while the specific mechanism remains to be elucidated. Circular RNAs (circRNAs) have been found to play critical roles in the tissue development and serve as biomarkers for various diseases. However, it remains unclear that the expression patterns of circRNAs and the roles of them played in recombinant NELL‐1‐induced osteogenesis of human adipose‐derived stem cells (hASCs). In this study, we performed RNA‐sequencing to investigate the expression profiles of circRNAs in recombinant NELL‐1‐induced osteogenic differentiation and identified two key circRNAs, namely circRFWD2 and circINO80. These two circRNAs were confirmed to be up‐regulated during recombinant NELL‐1‐induced osteogenesis, and knockdown of them affected the positive effect of NELL‐1 on osteogenesis. CircRFWD2 and circINO80 could interact with hsa‐miR‐6817‐5p, which could inhibit the osteogenesis. Silencing hsa‐miR‐6817‐5p could partially reverse the negative effect of si‐circRFWD2 and si‐circINO80 on the osteogenesis. Therefore, circRFWD2 and circINO80 could regulate the expression of hsa‐miR‐6817‐5p and influence the recombinant NELL‐1‐induced osteogenic differentiation of hASCs. It opens a new window to better understanding the effects of NELL‐1 on the osteogenic differentiation of hASCs and provides potential molecular targets and novel methods for bone regeneration efficiently and safely.
Collapse
Affiliation(s)
- Xinqi Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiao Cen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Temporomandibular Joint, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bo Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuwei Liao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhenxing Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Guanyin Zhu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jun Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
16
|
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: 3.8] [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
|
17
|
Sethi S, Debiec H, Madden B, Charlesworth MC, Morelle J, Gross L, Ravindran A, Buob D, Jadoul M, Fervenza FC, Ronco P. Neural epidermal growth factor-like 1 protein (NELL-1) associated membranous nephropathy. Kidney Int 2019; 97:163-174. [PMID: 31901340 DOI: 10.1016/j.kint.2019.09.014] [Citation(s) in RCA: 185] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/10/2019] [Accepted: 09/12/2019] [Indexed: 12/12/2022]
Abstract
Membranous nephropathy is characterized by deposition of immune complexes along the glomerular basement membrane. PLA2R and THSD7A are target antigens in 70% and 1-5% of primary membranous nephropathy cases, respectively. In the remaining cases, the target antigen is unknown. Here, laser microdissection of glomeruli followed by mass spectrometry was used to identify novel antigen(s) in PLA2R-negative membranous nephropathy. An initial pilot mass spectrometry study in 35 cases of PLA2R-negative membranous nephropathy showed high spectral counts for neural tissue encoding protein with EGF-like repeats, NELL-1, in six cases. Mass spectrometry failed to detect NELL-1 in 23 PLA2R-associated membranous nephropathy and 88 controls. NELL-1 was localized by immunohistochemistry, which showed bright granular glomerular basement membrane staining for NELL-1 in all six cases. Next, an additional 23 NELL-1 positive cases of membranous nephropathy were identified by immunohistochemistry in a discovery cohort of 91 PLA2R-negative membranous nephropathy cases, 14 were confirmed by mass spectrometry. Thus, 29 of 126 PLA2R-negative cases were positive for NELL-1. PLA2R-associated membranous nephropathy and controls stained negative for NELL-1. We then identified five NELL-1 positive cases of membranous nephropathy out of 84 PLA2R and THSD7A-negative cases in two validation cohorts from France and Belgium. By confocal microscopy, both IgG and NELL-1 co-localized to the glomerular basement membrane. Western blot analysis showed reactivity to NELL-1 in five available sera, but no reactivity in control sera. Clinical and biopsy findings of NELL-1 positive membranous nephropathy showed features of primary membranous nephropathy. Thus, a subset of membranous nephropathy is associated with accumulation and co-localization of NELL-1 and IgG along the glomerular basement membrane, and with anti-NELL-1 antibodies in the serum. Hence, NELL-1 defines a distinct type of primary membranous nephropathy.
Collapse
Affiliation(s)
- Sanjeev Sethi
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA.
| | - Hanna Debiec
- Sorbonne Université, Université Pierre et Marie Curie Paris 06, Paris, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche S 1155, Paris, France
| | - Benjamin Madden
- Medical Genome Facility, Proteomics Core, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Johann Morelle
- Division of Nephrology, Cliniques universitaires Saint-Luc, Brussels, Belgium; Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | - LouAnn Gross
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Aishwarya Ravindran
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - David Buob
- Sorbonne Université, Université Pierre et Marie Curie Paris 06, Paris, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche S 1155, Paris, France; Department of Pathology, Tenon Hospital, Paris, France
| | - Michel Jadoul
- Division of Nephrology, Cliniques universitaires Saint-Luc, Brussels, Belgium; Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | - Fernando C Fervenza
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Pierre Ronco
- Sorbonne Université, Université Pierre et Marie Curie Paris 06, Paris, France; Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche S 1155, Paris, France; Hôpital de Jour-Nephrology, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
18
|
Yilmaz E, Mihci E, Nur B, Alper ÖM, Taçoy Ş. Recent Advances in Craniosynostosis. Pediatr Neurol 2019; 99:7-15. [PMID: 31421914 DOI: 10.1016/j.pediatrneurol.2019.01.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 12/25/2018] [Accepted: 01/24/2019] [Indexed: 12/27/2022]
Abstract
Craniosynostosis is a pathologic craniofacial disorder and is defined as the premature fusion of one or more cranial (calvarial) sutures. Cranial sutures are fibrous joints consisting of nonossified mesenchymal cells that play an important role in the development of healthy craniofacial skeletons. Early fusion of these sutures results in incomplete brain development that may lead to complications of several severe medical conditions including seizures, brain damage, mental delay, complex deformities, strabismus, and visual and breathing problems. As a congenital disease, craniosynostosis has a heterogeneous origin that can be affected by genetic and epigenetic alterations, teratogens, and environmental factors and make the syndrome highly complex. To date, approximately 200 syndromes have been linked to craniosynostosis. In addition to being part of a syndrome, craniosynostosis can be nonsyndromic, formed without any additional anomalies. More than 50 nuclear genes that relate to craniosynostosis have been identified. Besides genetic factors, epigenetic factors like microRNAs and mechanical forces also play important roles in suture fusion. As craniosynostosis is a multifactorial disorder, evaluating the craniosynostosis syndrome requires and depends on all the information obtained from clinical findings, genetic analysis, epigenetic or environmental factors, or gene modulators. In this review, we will focus on embryologic and genetic studies, as well as epigenetic and environmental studies. We will discuss published studies and correlate the findings with unknown aspects of craniofacial disorders.
Collapse
Affiliation(s)
- Elanur Yilmaz
- Department of Medical Biology and Genetics, Akdeniz University Medical School, Antalya, Turkey
| | - Ercan Mihci
- Department of Pediatric Genetics, Akdeniz University Medical School, Antalya, Turkey
| | - Banu Nur
- Department of Pediatric Genetics, Akdeniz University Medical School, Antalya, Turkey
| | - Özgül M Alper
- Department of Medical Biology and Genetics, Akdeniz University Medical School, Antalya, Turkey.
| | - Şükran Taçoy
- Department of Pediatric Genetics, Akdeniz University Medical School, Antalya, Turkey
| |
Collapse
|
19
|
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: 0.8] [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
|
20
|
Han Q, Wang Q, Wu J, Li M, Fang Y, Zhu H, Wang X. Nell-1 promotes the neural-like differentiation of dental pulp cells. Biochem Biophys Res Commun 2019; 513:515-521. [PMID: 30979495 DOI: 10.1016/j.bbrc.2019.04.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 04/03/2019] [Indexed: 02/06/2023]
Abstract
Previous studies showed that Nel-like molecule-1 (Nell-1) can positively regulate odontoblastic differentiation and dentin formation. Intriguingly, our group found that Nell-1 is co-expressed with neural markers. The purpose of this study was to investigate whether Nell-1 protein plays a regulatory role in the differentiation of dental pulp cells into neural-like cells by in vivo and in vitro studies. The expression patterns of Nell-1 and dental pulp neural markers were observed by double immunofluorescence staining in normal dental pulp tissue sections of Wistar rat. Collagen sponge containing Nell-1 protein was added into the pulp cavity of rat molars in order to observe the expression patterns of neural markers in rat dental pulp repair and regeneration model by immunohistochemical staining. Moreover, human dental pulp stem cells (hDPSCs) were cultured, and different concentrations of Nell-1 protein were added for 12 h, 24 h, and 72h. The expression of neural markers was detected by using quantitative real-time polymerase chain reaction and Western blot. Nell-1 was co-expressed with neural markers including substance P (SP) and Nestin in rat dental pulp tissue. The expression of neural markers including SP, neuron-specific enolase (NSE), and Nestin was increased obviously in rat dental pulp tissues stimulated with Nell-1 protein. In cultured hDPSCs induced by Nell-1 protein, the expression of neural markers including glial fibrillary acidic protein (GFAP), Nestin, and β-III tubulin was increased. Nell-1 plays a positive role in inducing the differentiation of DPSCs into neural-like cells.
Collapse
Affiliation(s)
- Qi Han
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School and Hospital of Stomatology, Shandong University, 44-1Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Qiang Wang
- Jinan Stomatological Hospital, Jinan, 250001, Shandong, China
| | - Jiameng Wu
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School and Hospital of Stomatology, Shandong University, 44-1Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Mengyue Li
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School and Hospital of Stomatology, Shandong University, 44-1Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Yixuan Fang
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School and Hospital of Stomatology, Shandong University, 44-1Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Hongfan Zhu
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School and Hospital of Stomatology, Shandong University, 44-1Wenhuaxi Road, Jinan, 250012, Shandong, China
| | - Xiaoying Wang
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School and Hospital of Stomatology, Shandong University, 44-1Wenhuaxi Road, Jinan, 250012, Shandong, China.
| |
Collapse
|
21
|
Meyers CA, Sun Z, Chang L, Ding C, Lu A, Ting K, Pang S, James AW. Age dependent effects of NELL-1 isoforms on bone marrow stromal cells. J Orthop 2019; 16:175-178. [PMID: 30899146 PMCID: PMC6406628 DOI: 10.1016/j.jor.2019.02.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 02/17/2019] [Indexed: 10/27/2022] Open
Abstract
NELL-1 is an osteogenic protein first discovered to control ossification of the cranium. NELL-1 exists in at least two isoforms. The full-length NELL-1 contains 810 amino acid (aa) (NELL-1810), the N-terminal-truncated NELL-1 isoform contains 570 aa (NELL-1570). The differences in cellular effects between NELL-1 isoforms are not well understood. Methods: Here, BMSC were derived from adult or aged mice, followed by overexpression of NELL-1810 or NELL-1570. Cell morphology, proliferation, and gene expression were examined. Results/Conclusions: Overall, the proliferative effect of NELL-1570 was age dependent, showing prominent induction in adult but not aged mice.
Collapse
Affiliation(s)
| | - Zhibo Sun
- Department of Pathology, Johns Hopkins University, 21205, USA
| | - Leslie Chang
- Department of Pathology, Johns Hopkins University, 21205, USA
| | - Catherine Ding
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, USA
| | - Amy Lu
- Department of Pathology, Johns Hopkins University, 21205, USA
| | - Kang Ting
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, USA
| | - Shen Pang
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, 90095, USA
| | - Aaron W. James
- Department of Pathology, Johns Hopkins University, 21205, USA
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, 90095, USA
| |
Collapse
|
22
|
Qi H, Kim JK, Ha P, Chen X, Chen E, Chen Y, Li J, Pan HC, Yu M, Mohazeb Y, Azer S, Baik L, Kwak JH, Ting K, Zhang X, Hu M, Soo C. Inactivation of Nell-1 in Chondrocytes Significantly Impedes Appendicular Skeletogenesis. J Bone Miner Res 2019; 34:533-546. [PMID: 30352124 PMCID: PMC6677149 DOI: 10.1002/jbmr.3615] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 09/18/2018] [Accepted: 10/06/2018] [Indexed: 12/29/2022]
Abstract
NELL-1, an osteoinductive protein, has been shown to regulate skeletal ossification. Interestingly, an interstitial 11p14.1-p15.3 deletion involving the Nell-1 gene was recently reported in a patient with short stature and delayed fontanelle closure. Here we sought to define the role of Nell-1 in endochondral ossification by investigating Nell-1-specific inactivation in Col2α1-expressing cell lineages. Nell-1flox/flox ; Col2α1-Cre+ (Nell-1Col2α1 KO) mice were generated for comprehensive analysis. Nell-1Col2α1 KO mice were born alive but displayed subtle femoral length shortening. At 1 and 3 months postpartum, Nell-1 inactivation resulted in dwarfism and premature osteoporotic phenotypes. Specifically, Nell-1Col2α1 KO femurs and tibias exhibited significantly reduced length, bone mineral density (BMD), bone volume per tissue volume (BV/TV), trabecular number/thickness, cortical volume/thickness/density, and increased trabecular separation. The decreased bone formation rate revealed by dynamic histomorphometry was associated with altered numbers and/or function of osteoblasts and osteoclasts. Furthermore, longitudinal observations by in vivo micro-CT showed delayed and reduced mineralization at secondary ossification centers in mutants. Histologically, reduced staining intensities of Safranin O, Col-2, Col-10, and fewer BrdU-positive chondrocytes were observed in thinner Nell-1Col2α1 KO epiphyseal plates along with altered distribution and weaker expression level of Ihh, Patched-1, PTHrP, and PTHrP receptor. Primary Nell-1Col2α1 KO chondrocytes also exhibited decreased proliferation and differentiation, and its downregulated expression of the Ihh-PTHrP signaling molecules can be partially rescued by exogenous Nell-1 protein. Moreover, intranuclear Gli-1 protein and gene expression of the Gli-1 downstream target genes, Hip-1 and N-Myc, were also significantly decreased with Nell-1 inactivation. Notably, the rescue effects were diminished/reduced with application of Ihh signaling inhibitors, cyclopamine or GANT61. Taken together, these findings suggest that Nell-1 is a pivotal modulator of epiphyseal homeostasis and endochondral ossification. The cumulative chondrocyte-specific Nell-1 inactivation significantly impedes appendicular skeletogenesis resulting in dwarfism and premature osteoporosis through inhibiting Ihh signaling and predominantly altering the Ihh-PTHrP feedback loop. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Huichuan Qi
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, Changchun, Jilin, P. R. China
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Jong Kil Kim
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Pin Ha
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Xiaoyan Chen
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
- Department of Orthodontics, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Eric Chen
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Yao Chen
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Jiayi Li
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Hsin Chuan Pan
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Mengliu Yu
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
- Center of Stomatology, China-Japan Friendship Hospital, 2nd Yinghuayuan East Street, Chaoyang District, Beijing, P. R. China
| | - Yasamin Mohazeb
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Sophia Azer
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Lloyd Baik
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Jin Hee Kwak
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Kang Ting
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Xinli Zhang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Min Hu
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, Changchun, Jilin, P. R. China
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
23
|
Yamamoto N, Kashiwagi M, Ishihara M, Kojima T, Maturana AD, Kuroda S, Niimi T. Robo2 contains a cryptic binding site for neural EGFL-like (NELL) protein 1/2. J Biol Chem 2019; 294:4693-4703. [PMID: 30700556 DOI: 10.1074/jbc.ra118.005819] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 01/14/2019] [Indexed: 12/15/2022] Open
Abstract
The signaling pathways that are mediated by Slit ligands and their Roundabout (Robo) family of receptors play multifunctional roles in the development of the nervous system and other organs. A recent study identified neural epidermal growth factor-like (NEL)-like 2 (NELL2) as a novel ligand for Robo3. In this study, we carried out a comprehensive analysis of the interaction between NELL1 and the Robo family of receptors and demonstrated that Robo2 contains a cryptic binding site for both NELL1 and NELL2. NELL1/2 binds to the first fibronectin type III (FNIII) domain of Robo2 but not to intact Robo2. Mutation analysis revealed that several amino acids within the first FNIII domain are critical for NELL1 binding to Robo2 but not to Robo1. The Robo2 deletion mutants without the fourth immunoglobulin domain and single amino acid substitution mutants that can influence the architecture of the ectodomain facilitated binding to NELL1/2. Acidic conditions increased the binding affinity of Robo2 for NELL1. These results suggest that Robo2 functions as a receptor for NELL1/2, particularly under circumstances where Robo2 undergoes proteolytic digestion. If this is not the case, conformational changes of the ectodomain of Robo2 may unmask the binding site for NELL1/2.
Collapse
Affiliation(s)
- Naoka Yamamoto
- From the Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan and
| | - Manabu Kashiwagi
- From the Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan and
| | - Manami Ishihara
- From the Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan and
| | - Takaaki Kojima
- From the Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan and
| | - Andrés D Maturana
- From the Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan and
| | - Shun'ichi Kuroda
- the Institute of Scientific and Industrial Research, Osaka University, Osaka 567-0047, Japan
| | - Tomoaki Niimi
- From the Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan and
| |
Collapse
|
24
|
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: 21] [Impact Index Per Article: 3.5] [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
|
25
|
Optimizing Osteogenic Differentiation of Ovine Adipose-Derived Stem Cells by Osteogenic Induction Medium and FGFb, BMP2, or NELL1 In Vitro. Stem Cells Int 2018; 2018:9781393. [PMID: 30356449 PMCID: PMC6178511 DOI: 10.1155/2018/9781393] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 07/25/2018] [Accepted: 08/12/2018] [Indexed: 01/27/2023] Open
Abstract
Although adipose-derived stromal cells (ADSCs) have been a major focus as an alternative to autologous bone graft in orthopedic surgery, bone formation potential of ADSCs is not well known and cytokines as osteogenic inducers on ADSCs are being investigated. This study aimed at isolating ADSCs from ovine adipose tissue (AT) and optimizing osteogenic differentiation of ovine ADSCs (oADSC) by culture medium and growth factors. Four AT samples were harvested from two female ovine (Texel/Gotland breed), and oADSCs were isolated and analyzed by flow cytometry for surface markers CD29, CD44, CD31, and CD45. Osteogenic differentiation was made in vitro by seeding oADSCs in osteogenic induction medium (OIM) containing fibroblast growth factor basic (FGFb), bone morphogenetic protein 2 (BMP2), or NEL-like molecule 1 (NELL1) in 4 different dosages (1, 10, 50, and 100 ng/ml, respectively). Basic medium (DMEM) was used as control. Analysis was made after 14 days by Alizarin red staining (ARS) and quantification. This study successfully harvested AT from ovine and verified isolated cells for minimal criteria for adipose stromal cells which suggests a feasible method for isolation of oADSCs. OIM showed significantly higher ARS to basic medium, and FGFb 10 ng/ml revealed significantly higher ARS to OIM alone after 14 days.
Collapse
|
26
|
Abstract
Craniosynostosis is a common craniofacial birth defect. This review focusses on the advances that have been achieved through studying the pathogenesis of craniosynostosis using mouse models. Classic methods of gene targeting which generate individual gene knockout models have successfully identified numerous genes required for normal development of the skull bones and sutures. However, the study of syndromic craniosynostosis has largely benefited from the production of knockin models that precisely mimic human mutations. These have allowed the detailed investigation of downstream events at the cellular and molecular level following otherwise unpredictable gain-of-function effects. This has greatly enhanced our understanding of the pathogenesis of this disease and has the potential to translate into improvement of the clinical management of this condition in the future.
Collapse
Affiliation(s)
- Kevin K L Lee
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Philip Stanier
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Erwin Pauws
- UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
27
|
Zhao H, Qin X, Zhang Q, Zhang X, Lin J, Ting K, Chen F. Nell-1-ΔE, a novel transcript of Nell-1, inhibits cell migration by interacting with enolase-1. J Cell Biochem 2018; 119:5725-5733. [PMID: 29388706 DOI: 10.1002/jcb.26756] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 01/25/2018] [Indexed: 11/06/2022]
Abstract
NELL-1 is a secreted protein that was originally found to be upregulated in pathologically fusing and fused sutures in non-syndromic unilateral coronal synostosis patients. Apart from the ability of NELL-1 to promote osteogenesis in long and craniofacial bones, NELL-1 reportedly inhibits the formation of several benign and malignant tumors. We previously identified a novel transcript of Nell-1 that lacked a calcium-binding epidermal growth factor (EGF)-like domain compared with full-length Nell-1; this new transcript was named Nell-1-ΔE. Three obvious structural differences between these two isoforms were revealed by homology modeling. Furthermore, the recombinant Nell-1-ΔE protein, but not the full-length Nell-1 protein, inhibited cell migration in vitro. However, full-length Nell-1 and Nell-1-ΔE proteins were present in similar subcellular locations and displayed similar expression patterns in both the intracellular and extracellular spaces. The results from the co-immunoprecipitation and liquid chromatography/tandem mass spectrometry analyses using two cell lines demonstrated that Nell-1-ΔE but not full-length Nell-1 interacted with enolase-1 in the extracellular spaces of both cell lines. The results of wound healing assays using ENO-1-overexpressing cells treated with full-length Nell-1/Nell-1-ΔE suggested that Nell-1-ΔE inhibited cell migration by interacting with ENO-1. Our study indicated that the novel transcript Nell-1-ΔE, but not full-length Nell-1, might be a candidate tumor suppressor factor for basic research and clinical practice.
Collapse
Affiliation(s)
- Huaxiang Zhao
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, P. R. China
| | - Xueyan Qin
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, P. R. China
| | - Qian Zhang
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, P. R. China
| | - Xinli Zhang
- Dental and Craniofacial Research Institute, University of California, Los Angeles, California
| | - Jiuxiang Lin
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, P. R. China
| | - Kang Ting
- Dental and Craniofacial Research Institute, University of California, Los Angeles, California
| | - Feng Chen
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, P. R. China
| |
Collapse
|
28
|
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: 1.7] [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
|
29
|
Morse A, Schindeler A, McDonald MM, Kneissel M, Kramer I, Little DG. Sclerostin Antibody Augments the Anabolic Bone Formation Response in a Mouse Model of Mechanical Tibial Loading. J Bone Miner Res 2018; 33:486-498. [PMID: 29090474 DOI: 10.1002/jbmr.3330] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/11/2017] [Accepted: 10/29/2017] [Indexed: 12/18/2022]
Abstract
Decreased activity or expression of sclerostin, an endogenous inhibitor of Wnt/β-catenin signaling, results in increased bone formation and mass. Antibodies targeting and neutralizing sclerostin (Scl-Ab) have been shown to increase bone mass and reduce fracture risk. Sclerostin is also important in modulating the response of bone to changes in its biomechanical environment. However, the effects of Scl-Ab on mechanotransduction are unclear, and it was speculated that the loading response may be altered for individuals receiving Scl-Ab therapy. To address this, we carried out a 2-week study of tibial cyclic compressive loading on C57Bl/6 mice treated with vehicle or 100 mg/kg/wk Scl-Ab. Increases in bone volume, density, and dynamic bone formation were found with loading, and the anabolic response was further increased by the combination of load and Scl-Ab. To investigate the underlying mechanism, gene profiling by RNA sequencing (RNAseq) was performed on tibias isolated from mice from all four experimental groups. Major alterations in Wnt/β-catenin gene expression were found with tibial loading, however not with Scl-Ab treatment alone. Notably, the combination of load and Scl-Ab elicited a synergistic response from a number of specific Wnt-related and mechanotransduction factors. An unexpected finding was significant upregulation of factors in the Rho GTPase signaling pathway with combination treatment. In summary, combination therapy had a more profound anabolic response than either Scl-Ab or loading treatment alone. The Wnt/β-catenin and Rho GTPase pathways were implicated within bone mechanotransduction and support the concept that bone mechanotransduction is likely to encompass a number of interconnected signaling pathways. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Alyson Morse
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, Australia.,Discipline of Paediatrics and Child Health, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Aaron Schindeler
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, Australia.,Discipline of Paediatrics and Child Health, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Michelle M McDonald
- Bone Biology Program, The Garvan Institute of Medical Research, Darlinghurst, Australia
| | | | | | - David G Little
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, Australia.,Discipline of Paediatrics and Child Health, Sydney Medical School, University of Sydney, Sydney, Australia
| |
Collapse
|
30
|
Li C, Zheng Z, Jiang J, Jiang W, Lee K, Berthiaume EA, Chen EC, Culiat CT, Zhou YH, Zhang X, Ting K, Soo C. Neural EGFL-Like 1 Regulates Cartilage Maturation through Runt-Related Transcription Factor 3-Mediated Indian Hedgehog Signaling. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:392-403. [PMID: 29137952 PMCID: PMC5785559 DOI: 10.1016/j.ajpath.2017.09.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/02/2017] [Accepted: 09/26/2017] [Indexed: 02/05/2023]
Abstract
The pro-chondrogenic function of runt-related transcription factor 2 (Runx2) was previously considered to be dependent on direct binding with the promoter of Indian hedgehog (Ihh)-the major regulator of chondrocyte differentiation, proliferation, and maturation. The authors' previous studies identified neural EGFL like 1 (Nell-1) as a Runx2-responsive growth factor for chondrogenic differentiation and maturation. In this study, it was further revealed that the pro-chondrogenic activities of Nell-1 also rely on Ihh signaling, by showing: i) Nell-1 significantly elevated Ihh signal transduction; ii) Nell-1 deficiency markedly reduced Ihh activation in chondrocytes; and iii) Nell-1-stimulated chondrogenesis was significantly reduced by the specific hedgehog inhibitor cyclopamine. Importantly, the authors demonstrated that Nell-1-responsive Ihh signaling and chondrogenic differentiation extended to Runx2-/- models in vitro and in vivo. In Runx2-/- chondrocytes, Nell-1 stimulated the expression and signal transduction of Runx3, another transcription factor required for complete chondrogenic differentiation and maturation. Furthermore, knocking down Runx3 in Runx2-/- chondrocytes abolished Nell-1's stimulation of Ihh-associated molecule expression, which validates Runx3 as a major mediator of Nell-1-stimulated Ihh activation. For the first time, the Runx2→Nell-1→Runx3→Ihh signaling cascade during chondrogenic differentiation and maturation has been identified as an alternative, but critical, pathway for Runx2 to function as a pro-chondrogenic molecule via Nell-1.
Collapse
Affiliation(s)
- Chenshuang Li
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, California; Department of Orthodontics, Peking University, School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Zhong Zheng
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, California
| | - Jie Jiang
- UCLA Division of Plastic and Reconstructive Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, California
| | - Wenlu Jiang
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, California; State Key Laboratory of Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Kevin Lee
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, California
| | - Emily A Berthiaume
- David Geffen School of Medicine at UCLA, University of California, Los Angeles, California
| | - Eric C Chen
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, California
| | | | - Yan-Heng Zhou
- Department of Orthodontics, Peking University, School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Xinli Zhang
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, California
| | - Kang Ting
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, California; UCLA Division of Plastic and Reconstructive Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, California.
| | - Chia Soo
- UCLA Division of Plastic and Reconstructive Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, California.
| |
Collapse
|
31
|
Li C, Zheng Z, Zhang X, Asatrian G, Chen E, Song R, Culiat C, Ting K, Soo C. Nfatc1 Is a Functional Transcriptional Factor Mediating Nell-1-Induced Runx3 Upregulation in Chondrocytes. Int J Mol Sci 2018; 19:ijms19010168. [PMID: 29316655 PMCID: PMC5796117 DOI: 10.3390/ijms19010168] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 12/19/2017] [Accepted: 01/05/2018] [Indexed: 01/01/2023] Open
Abstract
Neural EGFL like 1 (Nell-1) is essential for chondrogenic differentiation, maturation, and regeneration. Our previous studies have demonstrated that Nell-1's pro-chondrogenic activities are predominantly reliant upon runt-related transcription factor 3 (Runx3)-mediated Indian hedgehog (Ihh) signaling. Here, we identify the nuclear factor of activated T-cells 1 (Nfatc1) as the key transcriptional factor mediating the Nell-1 → Runx3 signal transduction in chondrocytes. Using chromatin immunoprecipitation assay, we were able to determine that Nfatc1 binds to the -833--810 region of the Runx3-promoter in response to Nell-1 treatment. By revealing the Nell-1 → Nfatc1 → Runx3 → Ihh cascade, we demonstrate the involvement of Nfatc1, a nuclear factor of activated T-cells, in chondrogenesis, while providing innovative insights into developing a novel therapeutic strategy for cartilage regeneration and other chondrogenesis-related conditions.
Collapse
Affiliation(s)
- Chenshuang Li
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Zhong Zheng
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Xinli Zhang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Greg Asatrian
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Eric Chen
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Richard Song
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Cymbeline Culiat
- NellOne Therapeutics, Inc., 99 Midway Ln # E, Oak Ridge, TN 37830, USA.
| | - Kang Ting
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery and Department of Orthopaedic Surgery, the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
32
|
Bone Tissue Engineering Strategies in Co-Delivery of Bone Morphogenetic Protein-2 and Biochemical Signaling Factors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1078:233-244. [PMID: 30357626 DOI: 10.1007/978-981-13-0950-2_12] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Administration of bone morphogenetic protein-2 (BMP-2), which is commercially approved by the food and drug administration to damaged bone sites has been investigated for the purpose of bone tissue regeneration. BMP-2 can promote osteoblastic differentiation of mesenchymal stem cells as well as regeneration of bone formation in early phase. This review highlights various factors such as vitamin D, dexamethasone, platelet-derived growth factor, placental growth factor, BMP-7, and NEL-like protein-1 that enhance and stimulate angiogenesis, cell differentiation, and bone regeneration. These biochemical signals and growth factors (GFs) accelerate bone repair and remodeling either synergistically or individually. Delivery systems and scaffolds are used for sustained release of these cargo molecules and support at damaged bone sites. Compared with direct administration of BMP-2, current studies have demonstrated that a combination of multiple GFs and/or therapeutic chemical factors with delivery platforms synergistically facilitates bone regeneration. Therefore, in the future, multiple combinations of various GFs, chemicals, and materials could provide patients and surgeons with non-invasive treatment options without secondary surgery and pain. To the end, this review summarizes the biological functions and synergistic effects of dual administration modalities involving BMP-2 as well as recent developments in bone tissue engineering applications.
Collapse
|
33
|
Fahmy-Garcia S, van Driel M, Witte-Buoma J, Walles H, van Leeuwen JPTM, van Osch GJVM, Farrell E. NELL-1, HMGB1, and CCN2 Enhance Migration and Vasculogenesis, But Not Osteogenic Differentiation Compared to BMP2. Tissue Eng Part A 2017; 24:207-218. [PMID: 28463604 DOI: 10.1089/ten.tea.2016.0537] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Currently, autografts still represent the gold standard treatment for the repair of large bone defects. However, these are associated with donor-site morbidity and increased pain, cost, and recovery time. The ideal therapy would use biomaterials combined with bone growth factors to induce and instruct bone defect repair without the need to harvest patient tissue. In this line, bone morphogenetic proteins (BMPs) have been the most extensively used agents for clinical bone repair, but at supraphysiological doses that are not without risk. Because of the need to eliminate the risks of BMP2 use in vivo, we assessed the ability of three putative osteogenic factors, nel-like molecule type 1 (NELL-1), high mobility group box 1 (HMGB1), and CCN2, to enhance the essential processes for bone defect repair in vitro and compared them to BMP2. Although it has been reported that NELL-1, HMGB1, and CCN2 play a role in bone formation, less is known about the contribution of these proteins to the different events involved, such as cell migration, osteogenesis, and vasculogenesis. In this study, we investigated the effects of different doses of NELL-1, HMGB, CCN2, and BMP2 on these three processes as a model for the recruitment and differentiation of resident cells in the in vivo bone defect repair situation, using cells of human origin. Our data demonstrated that NELL-1, HMGB1, and CCN2 significantly induced mesenchymal stem cell migration (from 1.58-fold increase compared to control), but BMP2 did not. Interestingly, only BMP2 increased osteogenesis in marrow stromal cells, whereas it inhibited osteogenesis in preosteoblasts. Moreover, the four proteins studied promoted significantly endothelial cell migration, reaching a maximum of 2.4-fold increase compared to control, and induced formation of tube-like structures. NELL-1, HMGB1, and CCN2 had these effects at relatively low doses compared to BMP2. This work indicates that NELL-1, HMGB1, and CCN2 might enhance bone defect healing via the recruitment of endogenous cells and induction of vascularization and act via different processes than BMP2.
Collapse
Affiliation(s)
| | | | - Janneke Witte-Buoma
- 3 Department of Oral and Maxillofacial Surgery, Erasmus MC , Rotterdam, The Netherlands
| | - Heike Walles
- 4 Department Tissue Engineering and Regenerative Medicine, University Hospital Würzburg , Würzburg, Germany
| | | | - Gerjo J V M van Osch
- 1 Department of Orthopaedics, Erasmus MC , Rotterdam, The Netherlands .,5 Otorhinolaryngology Department, Erasmus MC, Rotterdam, The Netherlands
| | - Eric Farrell
- 3 Department of Oral and Maxillofacial Surgery, Erasmus MC , Rotterdam, The Netherlands
| |
Collapse
|
34
|
Abstract
BACKGROUND Nonhealing bone defects represent an immense biomedical burden. Despite recent advances in protein-based bone regeneration, safety concerns over bone morphogenetic protein-2 have prompted the search for alternative factors. Previously, the authors examined the additive/synergistic effects of hedgehog and Nel-like protein-1 (NELL-1) on the osteogenic differentiation of mesenchymal stem cells in vitro. In this study, the authors sought to leverage their previous findings by applying the combination of Smoothened agonist (SAG), hedgehog signal activator, and NELL-1 to an in vivo critical-size bone defect model. METHODS A 4-mm parietal bone defect was created in mixed-gender CD-1 mice. Treatment groups included control (n = 6), SAG (n = 7), NELL-1 (n = 7), and SAG plus NELL-1 (n = 7). A custom fabricated poly(lactic-co-glycolic acid) disk with hydroxyapatite coating was used as an osteoinductive scaffold. RESULTS Results at 4 and 8 weeks showed increased bone formation by micro-computed tomographic analyses with either stimulus alone (SAG or NELL-1), but significantly greater bone formation with both components combined (SAG plus NELL-1). This included greater bone healing scores and increased bone volume and bone thickness. Histologic analyses confirmed a significant increase in new bone formation with the combination therapy SAG plus NELL-1, accompanied by increased defect vascularization. CONCLUSIONS In summary, the authors' results suggest that combining the hedgehog signaling agonist SAG and NELL-1 has potential as a novel therapeutic strategy for the healing of critical-size bone defects. Future directions will include optimization of dosage and delivery strategy for an SAG and NELL-1 combination product.
Collapse
|
35
|
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: 2.3] [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
|
36
|
Discussion: Combining Smoothened Agonist and NEL-Like Protein-1 Enhances Bone Healing. Plast Reconstr Surg 2017; 139:1397-1398. [PMID: 28538564 DOI: 10.1097/prs.0000000000003368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
37
|
Song D, Zhang F, Reid RR, Ye J, Wei Q, Liao J, Zou Y, Fan J, Ma C, Hu X, Qu X, Chen L, Li L, Yu Y, Yu X, Zhang Z, Zhao C, Zeng Z, Zhang R, Yan S, Wu T, Wu X, Shu Y, Lei J, Li Y, Zhang W, Wang J, Lee MJ, Wolf JM, Huang D, He TC. BMP9 induces osteogenesis and adipogenesis in the immortalized human cranial suture progenitors from the patent sutures of craniosynostosis patients. J Cell Mol Med 2017; 21:2782-2795. [PMID: 28470873 PMCID: PMC5661262 DOI: 10.1111/jcmm.13193] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 03/09/2017] [Indexed: 02/05/2023] Open
Abstract
The cranial suture complex is a heterogeneous tissue consisting of osteogenic progenitor cells and mesenchymal stem cells (MSCs) from bone marrow and suture mesenchyme. The fusion of cranial sutures is a highly coordinated and tightly regulated process during development. Craniosynostosis is a congenital malformation caused by premature fusion of cranial sutures. While the progenitor cells derived from the cranial suture complex should prove valuable for studying the molecular mechanisms underlying suture development and pathogenic premature suture fusion, primary human cranial suture progenitors (SuPs) have limited life span and gradually lose osteoblastic ability over passages. To overcome technical challenges in maintaining sufficient and long-term culture of SuPs for suture biology studies, we establish and characterize the reversibly immortalized human cranial suture progenitors (iSuPs). Using a reversible immortalization system expressing SV40 T flanked with FRT sites, we demonstrate that primary human suture progenitor cells derived from the patent sutures of craniosynostosis patients can be efficiently immortalized. The iSuPs maintain long-term proliferative activity, express most of the consensus MSC markers and can differentiate into osteogenic and adipogenic lineages upon BMP9 stimulation in vitro and in vivo. The removal of SV40 T antigen by FLP recombinase results in a decrease in cell proliferation and an increase in the endogenous osteogenic and adipogenic capability in the iSuPs. Therefore, the iSuPs should be a valuable resource to study suture development, intramembranous ossification and the pathogenesis of craniosynostosis, as well as to explore cranial bone tissue engineering.
Collapse
Affiliation(s)
- Dongzhe Song
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Fugui Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Russell R Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Department of Surgery, Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL, USA
| | - Jixing Ye
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Department of Biomedical Engineering, School of Bioengineering, Chongqing University, Chongqing, China
| | - Qiang Wei
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Junyi Liao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Yulong Zou
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Jiaming Fan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Chao Ma
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Departments of Neurosurgery and Otolaryngology-Head & Neck Surgery, the Affiliated Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xue Hu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Xiangyang Qu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Liqun Chen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Li Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Department of Biomedical Engineering, School of Bioengineering, Chongqing University, Chongqing, China
| | - Yichun Yu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Department of Emergency Medicine, Beijing Hospital, Beijing, China
| | - Xinyi Yu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Zhicai Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Department of Orthopaedic Surgery, Union Hospital of Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Chen Zhao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Zongyue Zeng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Ruyi Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Shujuan Yan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Tingting Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Department of Biomedical Engineering, School of Bioengineering, Chongqing University, Chongqing, China
| | - Xingye Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Yi Shu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Jiayan Lei
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Yasha Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Wenwen Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Department of Laboratory Medicine and Clinical Diagnostics, the Affiliated Yantai Hospital, Binzhou Medical University, Yantai, China
| | - Jia Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine and the Affiliated Hospitals of Chongqing Medical University, Chongqing, China
| | - Michael J Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Jennifer Moriatis Wolf
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Dingming Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| |
Collapse
|
38
|
Li C, Jiang J, Zheng Z, Lee KS, Zhou Y, Chen E, Culiat CT, Qiao Y, Chen X, Ting K, Zhang X, Soo C. Neural EGFL-Like 1 Is a Downstream Regulator of Runt-Related Transcription Factor 2 in Chondrogenic Differentiation and Maturation. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:963-972. [PMID: 28302495 PMCID: PMC5417045 DOI: 10.1016/j.ajpath.2016.12.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 11/21/2016] [Accepted: 12/22/2016] [Indexed: 10/20/2022]
Abstract
Recent studies indicate that neural EGFL-like 1 (Nell-1), a secretive extracellular matrix molecule, is involved in chondrogenic differentiation. Herein, we demonstrated that Nell-1 serves as a key downstream target of runt-related transcription factor 2 (Runx2), a central regulator of chondrogenesis. Unlike in osteoblast lineage cells where Nell-1 and Runx2 demonstrate mutual regulation, further studies in chondrocytes revealed that Runx2 tightly regulates the expression of Nell-1; however, Nell-1 does not alter the expression of Runx2. More important, Nell-1 administration partially restored Runx2 deficiency-induced impairment of chondrocyte differentiation and maturation in vitro, ex vivo, and in vivo. Mechanistically, although the expression of Nell-1 is highly reliant on Runx2, the prochondrogenic function of Nell-1 persisted in Runx2-/- scenarios. The biopotency of Nell-1 is independent of the nuclear import and DNA binding functions of Runx2 during chondrogenesis. Nell-1 is a key functional mediator of chondrogenesis, thus opening up new possibilities for the application of Nell-1 in cartilage regeneration.
Collapse
Affiliation(s)
- Chenshuang Li
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, California; Department of Orthodontics, Peking University, School and Hospital of Stomatology, Beijing, China
| | - Jie Jiang
- UCLA Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Zhong Zheng
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Kevin S Lee
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Yanheng Zhou
- Department of Orthodontics, Peking University, School and Hospital of Stomatology, Beijing, China
| | - Eric Chen
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | | | - Yiqiang Qiao
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, California; Department of Orthodontics, School of Stomatology, Zhengzhou University, Zhengzhou, China
| | - Xuepeng Chen
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, California; Department of Orthodontics, Hospital of Stomatology, Zhejiang University, Hangzhou, China
| | - Kang Ting
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, California; UCLA Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Xinli Zhang
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, California.
| | - Chia Soo
- UCLA Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
39
|
Lattanzi W, Barba M, Di Pietro L, Boyadjiev SA. Genetic advances in craniosynostosis. Am J Med Genet A 2017; 173:1406-1429. [PMID: 28160402 DOI: 10.1002/ajmg.a.38159] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 12/30/2016] [Accepted: 01/06/2017] [Indexed: 12/22/2022]
Abstract
Craniosynostosis, the premature ossification of one or more skull sutures, is a clinically and genetically heterogeneous congenital anomaly affecting approximately one in 2,500 live births. In most cases, it occurs as an isolated congenital anomaly, that is, nonsyndromic craniosynostosis (NCS), the genetic, and environmental causes of which remain largely unknown. Recent data suggest that, at least some of the midline NCS cases may be explained by two loci inheritance. In approximately 25-30% of patients, craniosynostosis presents as a feature of a genetic syndrome due to chromosomal defects or mutations in genes within interconnected signaling pathways. The aim of this review is to provide a detailed and comprehensive update on the genetic and environmental factors associated with NCS, integrating the scientific findings achieved during the last decade. Focus on the neurodevelopmental, imaging, and treatment aspects of NCS is also provided.
Collapse
Affiliation(s)
- Wanda Lattanzi
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Latium Musculoskeletal Tıssue Bank, Rome, Italy
| | - Marta Barba
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Lorena Di Pietro
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Simeon A Boyadjiev
- Division of Genomic Medicine, Department of Pediatrics, Davis Medical Center, University of California, Sacramento, California
| |
Collapse
|
40
|
Dateki S, Watanabe S, Kinoshita F, Yoshiura KI, Moriuchi H. Identification of 11p14.1-p15.3 deletion probably associated with short stature, relative macrocephaly, and delayed closure of the fontanelles. Am J Med Genet A 2016; 173:217-220. [PMID: 27662520 DOI: 10.1002/ajmg.a.37978] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 08/29/2016] [Indexed: 01/07/2023]
Abstract
We herein report a de novo hemizygous 9.2-Mb interstitial deletion of chromosome 11p14.1-15.3 in a 3-year-old Japanese girl with short stature, relative macrocephaly, and delayed closure of cranial fontanelles and sutures. She did not show either any motor or mental development delay. This deletion involves 25 genes including NELL1. The loss of the Nell1 function leads to skeletal defects in the cranial vault and vertebral column, and overexpression of Nell1 causes craniosynostosis in mice. These results imply that short stature and an abnormality of membranous ossification could be explained by haploinsufficiency of NELL1 on 11p14.1-p15.3. Further studies are needed to clarify the phenotype in patients with an 11p14.1-15.3 deletion and the pathogenesis of NELL1. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sumito Dateki
- Department of Pediatrics, Nagasaki University Hospital, Nagasaki, Japan
| | - Satoshi Watanabe
- Department of Pediatrics, Nagasaki University Hospital, Nagasaki, Japan.,Department of Human Genetics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Fumiko Kinoshita
- Department of Pediatrics, Nagasaki University Hospital, Nagasaki, Japan
| | - Koh-Ichiro Yoshiura
- Department of Human Genetics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hiroyuki Moriuchi
- Department of Pediatrics, Nagasaki University Hospital, Nagasaki, Japan
| |
Collapse
|
41
|
Cifuentes-Mendiola S, Pérez-Martínez I, Muñoz-Saavedra Á, Torres-Contreras J, García-Hernández A. Clinical applications of molecular basis for Craniosynostosis. A narrative review. JOURNAL OF ORAL RESEARCH 2016. [DOI: 10.17126/joralres.2016.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
42
|
Li CS, Yang P, Ting K, Aghaloo T, Lee S, Zhang Y, Khalilinejad K, Murphy MC, Pan HC, Zhang X, Wu B, Zhou YH, Zhao Z, Zheng Z, Soo C. Fibromodulin reprogrammed cells: A novel cell source for bone regeneration. Biomaterials 2016; 83:194-206. [PMID: 26774565 DOI: 10.1016/j.biomaterials.2016.01.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 12/27/2015] [Accepted: 01/01/2016] [Indexed: 02/05/2023]
Abstract
Pluripotent or multipotent cell-based therapeutics are vital for skeletal reconstruction in non-healing critical-sized defects since the local endogenous progenitor cells are not often adequate to restore tissue continuity or function. However, currently available cell-based regenerative strategies are hindered by numerous obstacles including inadequate cell availability, painful and invasive cell-harvesting procedures, and tumorigenesis. Previously, we established a novel platform technology for inducing a quiescent stem cell-like stage using only a single extracellular proteoglycan, fibromodulin (FMOD), circumventing gene transduction. In this study, we further purified and significantly increased the reprogramming rate of the yield multipotent FMOD reprogrammed (FReP) cells. We also exposed the 'molecular blueprint' of FReP cell osteogenic differentiation by gene profiling. Radiographic analysis showed that implantation of FReP cells into a critical-sized SCID mouse calvarial defect, contributed to the robust osteogenic capability of FReP cells in a challenging clinically relevant traumatic scenario in vivo. The persistence, engraftment, and osteogenesis of transplanted FReP cells without tumorigenesis in vivo were confirmed by histological and immunohistochemical staining. Taken together, we have provided an extended potency, safety, and molecular profile of FReP cell-based bone regeneration. Therefore, FReP cells present a high potential for cellular and gene therapy products for bone regeneration.
Collapse
Affiliation(s)
- Chen-Shuang Li
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA; Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Pu Yang
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Kang Ting
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Tara Aghaloo
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Soonchul Lee
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA; Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University, Gyeonggi-do, 463-712, South Korea
| | - Yulong Zhang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Kambiz Khalilinejad
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Maxwell C Murphy
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Hsin Chuan Pan
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Xinli Zhang
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Benjamin Wu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yan-Heng Zhou
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Zhong Zheng
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Chia Soo
- UCLA Division of Plastic Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
43
|
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: 6.5] [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
|
44
|
Liu M, Wang Q, Tang R, Cao R, Wang X. Nel-like Molecule 1 Contributes to the Odontoblastic Differentiation of Human Dental Pulp Cells. J Endod 2016; 42:95-100. [DOI: 10.1016/j.joen.2015.08.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 08/16/2015] [Accepted: 08/27/2015] [Indexed: 10/22/2022]
|
45
|
Pang S, Shen J, Liu Y, Chen F, Zheng Z, James AW, Hsu CY, Zhang H, Lee KS, Wang C, Li C, Chen X, Jia H, Zhang X, Soo C, Ting K. Proliferation and osteogenic differentiation of mesenchymal stem cells induced by a short isoform of NELL-1. Stem Cells 2015; 33:904-15. [PMID: 25376942 DOI: 10.1002/stem.1884] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 09/19/2014] [Accepted: 09/27/2014] [Indexed: 01/11/2023]
Abstract
Neural epidermal growth factor-like (NEL)-like protein 1 (NELL-1) has been identified as an osteoinductive differentiation factor that promotes mesenchymal stem cell (MSC) osteogenic differentiation. In addition to full-length NELL-1, there are several NELL-1-related transcripts reported. We used rapid amplification of cDNA ends to recover potential cDNA of NELL-1 isoforms. A NELL-1 isoform with the N-terminal 240 amino acid (aa) residues truncated was identified. While full-length NELL-1 that contains 810 aa residues (NELL-1810 ) plays an important role in embryologic skeletal development, the N-terminal-truncated NELL-1 isoform (NELL-1570 ) was expressed postnatally. Similar to NELL-1810 , NELL-1570 induced MSC osteogenic differentiation. In addition, NELL-1570 significantly stimulated MSC proliferation in multiple MSC-like populations such as murine C3H10T1/2 MSC cell line, mouse primary MSCs, and perivascular stem cells, which is a type of stem cells proposed as the perivascular origin of MSCs. In contrast, NELL-1810 demonstrated only limited stimulation of MSC proliferation. Similar to NELL-1810 , NELL-1570 was found to be secreted from host cells. Both NELL-1570 expression lentiviral vector and column-purified recombinant protein NELL-1570 demonstrated almost identical effects in MSC proliferation and osteogenic differentiation, suggesting that NELL-1570 may function as a pro-osteogenic growth factor. In vivo, NELL-1570 induced significant calvarial defect regeneration accompanied by increased cell proliferation. Thus, NELL-1570 has the potential to be used for cell-based or hormone-based therapy of bone regeneration.
Collapse
Affiliation(s)
- Shen Pang
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Takahashi K, Imai A, Iijima M, Yoshimoto N, Maturana AD, Kuroda S, Niimi T. Mapping the heparin-binding site of the osteoinductive protein NELL1 by site-directed mutagenesis. FEBS Lett 2015; 589:4026-32. [PMID: 26627376 DOI: 10.1016/j.febslet.2015.11.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 10/28/2015] [Accepted: 11/18/2015] [Indexed: 01/13/2023]
Abstract
Neural epidermal growth factor-like (NEL)-like 1 (NELL1) is a secretory osteogenic protein comprising an N-terminal thrombospondin-1-like (TSPN) domain, four von Willebrand factor type C domains, and six epidermal growth factor-like repeats. NELL1 shows heparin-binding activity; however, the biological significance remains to be explored. In this report, we demonstrate that NELL1 binds to cell surface proteoglycans through its TSPN domain. Major heparin-binding sites were identified on the three-dimensional structural model of the TSPN domain of NELL1. Mutant analysis of the heparin-binding sites indicated that the heparin-binding activity of the TSPN domain is involved in interaction of NELL1 with cell surface proteoglycans.
Collapse
Affiliation(s)
- Kaneyoshi Takahashi
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Arisa Imai
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Masumi Iijima
- The Institute of Scientific and Industrial Research, Osaka University, Osaka, Japan
| | - Nobuo Yoshimoto
- The Institute of Scientific and Industrial Research, Osaka University, Osaka, Japan
| | - Andrés D Maturana
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Shun'ichi Kuroda
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan; The Institute of Scientific and Industrial Research, Osaka University, Osaka, Japan
| | - Tomoaki Niimi
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan.
| |
Collapse
|
47
|
The Development of the Calvarial Bones and Sutures and the Pathophysiology of Craniosynostosis. Curr Top Dev Biol 2015; 115:131-56. [PMID: 26589924 DOI: 10.1016/bs.ctdb.2015.07.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The skull vault is a complex, exquisitely patterned structure that plays a variety of key roles in vertebrate life, ranging from the acquisition of food to the support of the sense organs for hearing, smell, sight, and taste. During its development, it must meet the dual challenges of protecting the brain and accommodating its growth. The bones and sutures of the skull vault are derived from cranial neural crest and head mesoderm. The frontal and parietal bones develop from osteogenic rudiments in the supraorbital ridge. The coronal suture develops from a group of Shh-responsive cells in the head mesoderm that are collocated, with the osteogenic precursors, in the supraorbital ridge. The osteogenic rudiments and the prospective coronal suture expand apically by cell migration. A number of congenital disorders affect the skull vault. Prominent among these is craniosynostosis, the fusion of the bones at the sutures. Analysis of the pathophysiology underling craniosynostosis has identified a variety of cellular mechanisms, mediated by a range of signaling pathways and effector transcription factors. These cellular mechanisms include loss of boundary integrity, altered sutural cell specification in embryos, and loss of a suture stem cell population in adults. Future work making use of genome-wide transcriptomic approaches will address the deep structure of regulatory interactions and cellular processes that unify these seemingly diverse mechanisms.
Collapse
|
48
|
Park SS, Beyer RP, Smyth MD, Clarke CM, Timms AE, Bammler TK, Stamper BD, Mecham BH, Gustafson JA, Cunningham ML. Osteoblast differentiation profiles define sex specific gene expression patterns in craniosynostosis. Bone 2015; 76:169-76. [PMID: 25753363 PMCID: PMC4546839 DOI: 10.1016/j.bone.2015.03.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 02/18/2015] [Accepted: 03/01/2015] [Indexed: 01/25/2023]
Abstract
Single suture craniosynostosis (SSC) is the premature fusion of one calvarial suture and occurs in 1-1700-2500 live births. Congenital fusion of either the sagittal, metopic, or coronal sutures represents 95% of all cases of SSC. Sagittal and metopic synostosis have a male preponderance (3:1) while premature fusion of the coronal suture has a female preponderance (2:1). Although environmental and genetic factors contribute to SSC, the etiology of the majority of SSC cases remains unclear. In this study, 227 primary calvarial osteoblast cell lines from patients with coronal, metopic, or sagittal synostosis and unaffected controls were established and assayed for ALP activity and BrdU incorporation (n = 226) as respective measures of early stage osteoblast differentiation and proliferation. Primary osteoblast cell lines from individuals with sagittal synostosis demonstrated higher levels of ALP activity and reduced proliferation when compared to control lines. In order to address the sex differences in SSC types, the data was further stratified by sex. Osteoblasts from males and females with sagittal synostosis as well as males with metopic synostosis demonstrated higher levels of ALP activity when compared to sex matched controls, and males with sagittal or metopic synostosis demonstrated reduced levels of proliferation. In order to elucidate genes and pathways involved in these observed phenotypes, correlation analyses comparing ALP activity and proliferation to global gene expression was performed. Transcripts related to osteoblast differentiation were identified both differentially up and downregulated, correlated with ALP activity when compared to controls, and demonstrated a striking sex specific gene expression pattern. These data support that the dysregulation of osteoblast differentiation plays a role in the development of SSC and that genetic factors contribute to the observed sex related differences.
Collapse
Affiliation(s)
- Sarah S Park
- Seattle Children's Research Institute, Center for Developmental Biology and Regenerative Medicine, Seattle, WA, USA
| | - Richard P Beyer
- University of Washington, Center for Ecogenetics and Environmental Health, Seattle, WA, USA
| | - Matthew D Smyth
- Washington University, Department of Neurosurgery and St. Louis Children's Hospital, St. Louis, MO, USA
| | - Christine M Clarke
- Seattle Children's Research Institute, Center for Developmental Biology and Regenerative Medicine, Seattle, WA, USA
| | - Andrew E Timms
- Seattle Children's Research Institute, Center for Developmental Biology and Regenerative Medicine, Seattle, WA, USA
| | - Theo K Bammler
- University of Washington, Center for Ecogenetics and Environmental Health, Seattle, WA, USA
| | | | | | - Jennifer A Gustafson
- Seattle Children's Research Institute, Center for Developmental Biology and Regenerative Medicine, Seattle, WA, USA
| | - Michael L Cunningham
- Seattle Children's Research Institute, Center for Developmental Biology and Regenerative Medicine, Seattle, WA, USA; Seattle Children's Craniofacial Center, Seattle, WA, USA.
| |
Collapse
|
49
|
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: 9.6] [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
|
50
|
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
|