1
|
Li Z, Tian Y. Role of NEL‑like molecule‑1 in osteogenesis/chondrogenesis (Review). Int J Mol Med 2025; 55:5. [PMID: 39450541 PMCID: PMC11537270 DOI: 10.3892/ijmm.2024.5446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/09/2024] [Indexed: 10/26/2024] Open
Abstract
A dynamic balance exists between osteogenesis and osteoclastogenesis in bone tissue, which can lead to several bone diseases, such as osteoporosis, osteoarthritis, bone necrosis and bone defects, in cases of insufficient osteogenesis or excessive osteoclastogenesis. NEL‑like molecule‑1 (NELL‑1) was first discovered in 1999 as an osteogenic factor that can prevent or treat bone diseases by increasing osteogenic levels. To date, research has identified multiple signaling pathways involved in improving osteogenic levels. Furthermore, to apply NELL‑1 in clinical practice, researchers have optimized its osteogenic effect by combining it with other molecules, changing its molecular structure and performing bone tissue engineering. Currently, research on NELL‑1 is gaining increasing attention. In the near future, it will definitely be applied in clinical practice to eliminate diseases. Thus, the present study provides a comprehensive review of NELL‑1 in enhancing osteogenic levels from the perspectives of the molecular mechanism, interactions with other molecules/cells, molecular‑level changes, applications in bone tissue engineering and its expression in tumors, providing a solid theoretical basis for its clinical application.
Collapse
Affiliation(s)
- Zihan Li
- Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yihao Tian
- Department of Pathology, Beifang Hospital of China Medical University, General Hospital of Northern Theater Command, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
2
|
Bidkhori M, Akbarzadeh M, Fahimfar N, Jahangiri M, Seddiq S, Larijani B, Nabipour I, Mohammad Amoli M, Panahi N, Dehghan A, Holakouie-Naieni K, Ostovar A. Neural EGFL like 1 as a novel gene for Trabecular Bone Score in older adults: The Bushehr Elderly Health (BEH) program. PLoS One 2024; 19:e0309401. [PMID: 39255297 PMCID: PMC11386414 DOI: 10.1371/journal.pone.0309401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/13/2024] [Indexed: 09/12/2024] Open
Abstract
Neural EGFL like 1 (NELL-1), is a secreted glycoprotein and stimulates osteogenic cell differentiation and bone mineralization. This study aimed to explore the relationship between NELL-1 and Trabecular Bone Score (TBS) as a novel tool for the evaluation of osteoporosis in an elderly population-based cohort study in Iran. A single-locus analysis was performed on TBS using data from 2,071 participants in the Bushehr Elderly Health (BEH) Program. The study investigated 376 independent single nucleotide polymorphisms (SNPs) within the NELL-1 on chromosome 11p15.1. The association between SNPs and the mean TBS L1 to L4 was analyzed through an additive model. Significant variants in the additive model (PFDR<0.05) were further examined within dominant, recessive, over-dominant, and co-dominant models. Multiple linear regression was employed to assess the relationship between the genetic risk score (GRS) derived from significant SNPs and TBS. Three SNPs within the NELL-1 showed a statistically significant association with TBS after adjusting for age and sex. The associations for rs1901945 (β = 0.013, PFDR = 0.0007), rs1584851 (β = -0.011, PFDR = 0.0003), and rs58028601 (β = 0.011, PFDR = 0.0003) were significant in the additive model. Additionally, significant results were observed for rs1901945 and rs58028601 in the dominant model (P<0.05). The GRS showed a statistically significant relationship with TBS, considering adjustments for age, sex, Body Mass Index, type 2 diabetes, and smoking (β = 0.077, P = 1.7×10-5). This study highlights the association of NELL-1 with TBS, underscoring its potential as a candidate for further research and personalized medicine concerning the impact of this gene on bone quality.
Collapse
Affiliation(s)
- Mohammad Bidkhori
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Osteoporosis Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Akbarzadeh
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Noushin Fahimfar
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Osteoporosis Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Jahangiri
- Department of Biostatistics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sahar Seddiq
- Osteoporosis Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Iraj Nabipour
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mahsa Mohammad Amoli
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular -Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Nekoo Panahi
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular -Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Dehghan
- Department of Biostatistics and Epidemiology, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, United Kingdom
| | - Kourosh Holakouie-Naieni
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Afshin Ostovar
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Osteoporosis Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Liu N, Sun W, Gao W, Yan S, Yang C, Zhang J, Ni B, Zhang L, Zang J, Zhang S, Xu D. CD300e: Emerging role and mechanism as an immune-activating receptor. Int Immunopharmacol 2024; 133:112055. [PMID: 38677094 DOI: 10.1016/j.intimp.2024.112055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/06/2024] [Accepted: 04/07/2024] [Indexed: 04/29/2024]
Abstract
As a transmembrane protein, CD300e is primarily expressed in myeloid cells. It belongs to the CD300 glycoprotein family, functioning as an immune-activating receptor. Dysfunction of CD300e has been suggested in many diseases, such as infections, immune disorders, obesity, and diabetes, signifying its potential as a key biomarker for disease diagnosis and treatment. This review is aimed to explore the roles and potential mechanisms of CD300e in regulating oxidative stress, immune cell activation, tissue damage and repair, and lipid metabolism, shedding light on its role as a diagnostic marker or a therapeutic target, particularly for infections and autoimmune disorders.
Collapse
Affiliation(s)
- Na Liu
- Central Laboratory of the First Affiliated Hospital & the First Clinical College, Shandong Second Medical University, Weifang 261000, China
| | - Wenchang Sun
- Central Laboratory of the First Affiliated Hospital & the First Clinical College, Shandong Second Medical University, Weifang 261000, China
| | - Weixing Gao
- Office of the First Clinical Medical College, Shandong Second Medical University, Weifang 261000, China
| | - Shushan Yan
- Department of Colorectal and Anal Surgery of the First Affiliated Hospital & the First Clinical College, Shandong Second Medical University, Weifang 261000, China
| | - Chunjuan Yang
- Central Laboratory of the First Affiliated Hospital & the First Clinical College, Shandong Second Medical University, Weifang 261000, China; Department of Rheumatology of the Affiliated Hospital, Shandong Second Medical University, Weifang, 261053, China
| | - Jin Zhang
- Department of Colorectal and Anal Surgery of the First Affiliated Hospital & the First Clinical College, Shandong Second Medical University, Weifang 261000, China
| | - Biao Ni
- Central Laboratory of the First Affiliated Hospital & the First Clinical College, Shandong Second Medical University, Weifang 261000, China
| | - Lili Zhang
- Central Laboratory of the First Affiliated Hospital & the First Clinical College, Shandong Second Medical University, Weifang 261000, China
| | - Jie Zang
- Central Laboratory of the First Affiliated Hospital & the First Clinical College, Shandong Second Medical University, Weifang 261000, China
| | - Sue Zhang
- Department of Anesthesiology, Weifang People's Hospital, Weifang 261000, China.
| | - Donghua Xu
- Central Laboratory of the First Affiliated Hospital & the First Clinical College, Shandong Second Medical University, Weifang 261000, China; Department of Rheumatology of the Affiliated Hospital, Shandong Second Medical University, Weifang, 261053, China; Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
4
|
Andeen NK, Kung VL, Avasare RS. NELL1 membranous nephropathy: clinical associations provide mechanistic clues. FRONTIERS IN NEPHROLOGY 2024; 4:1323432. [PMID: 38596642 PMCID: PMC11002321 DOI: 10.3389/fneph.2024.1323432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 03/04/2024] [Indexed: 04/11/2024]
Abstract
Neural epidermal growth factor-like 1 (NELL1) membranous nephropathy (MN) is notable for its segmental deposit distribution, IgG1 dominant deposits, and comparatively high rate of spontaneous remission. It has been associated with a variety of exposures and secondary conditions, specifically use of thiol-containing medications - including lipoic acid, bucillamine, and tiopronin - as well as traditional indigenous medications (TIM) particularly those with high mercury content, and non-steroid anti-inflammatory drugs (NSAIDs). Malignancies, graft vs. host disease (GVHD), infection, and autoimmune conditions have also been associated with NELL1 MN. Herein, we provide a detailed summary of the clinicopathologic features of NELL1 and associations with underlying conditions, with a focus on treatment and outcomes. Rare cases of dual NELL1 and phospholipase A2 receptor (PLA2R) positive MN are reviewed. Genome-wide association study of NELL1, role of NELL1 in other physiologic and pathologic processes, and connection between NELL1 MN and malignancy with relevance of NELL1 tumor staining are examined. Finally, relationships and potential disease mechanisms of thiol- and mercury- associated NELL1 MN are discussed.
Collapse
Affiliation(s)
- Nicole K. Andeen
- Department of Pathology and Laboratory Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Vanderlene L. Kung
- Department of Pathology and Laboratory Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Rupali S. Avasare
- Department of Medicine, Division of Nephrology and Hypertension, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
5
|
Ha P, Kwak JH, Zhang Y, Shi J, Tran L, Liu TP, Pan HC, Lee S, Kim JK, Chen E, Shirazi-Fard Y, Stodieck LS, Lin A, Zheng Z, Dong SN, Zhang X, Wu BM, Ting K, Soo C. Bisphosphonate conjugation enhances the bone-specificity of NELL-1-based systemic therapy for spaceflight-induced bone loss in mice. NPJ Microgravity 2023; 9:75. [PMID: 37723136 PMCID: PMC10507033 DOI: 10.1038/s41526-023-00319-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/18/2023] [Indexed: 09/20/2023] Open
Abstract
Microgravity-induced bone loss results in a 1% bone mineral density loss monthly and can be a mission critical factor in long-duration spaceflight. Biomolecular therapies with dual osteogenic and anti-resorptive functions are promising for treating extreme osteoporosis. We previously confirmed that NELL-like molecule-1 (NELL-1) is crucial for bone density maintenance. We further PEGylated NELL-1 (NELL-polyethylene glycol, or NELL-PEG) to increase systemic delivery half-life from 5.5 to 15.5 h. In this study, we used a bio-inert bisphosphonate (BP) moiety to chemically engineer NELL-PEG into BP-NELL-PEG and specifically target bone tissues. We found conjugation with BP improved hydroxyapatite (HA) binding and protein stability of NELL-PEG while preserving NELL-1's osteogenicity in vitro. Furthermore, BP-NELL-PEG showed superior in vivo bone specificity without observable pathology in liver, spleen, lungs, brain, heart, muscles, or ovaries of mice. Finally, we tested BP-NELL-PEG through spaceflight exposure onboard the International Space Station (ISS) at maximal animal capacity (n = 40) in a long-term (9 week) osteoporosis therapeutic study and found that BP-NELL-PEG significantly increased bone formation in flight and ground control mice without obvious adverse health effects. Our results highlight BP-NELL-PEG as a promising therapeutic to mitigate extreme bone loss from long-duration microgravity exposure and musculoskeletal degeneration on Earth, especially when resistance training is not possible due to incapacity (e.g., bone fracture, stroke).
Collapse
Affiliation(s)
- Pin Ha
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jin Hee Kwak
- Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90089, USA
| | - Yulong Zhang
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Forsyth Institute, Cambridge, MA, 02142, USA
| | - Jiayu Shi
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Luan Tran
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Timothy Pan Liu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Hsin-Chuan Pan
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Samantha Lee
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jong Kil Kim
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Eric Chen
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yasaman Shirazi-Fard
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - Louis S Stodieck
- BioServe Space Technologies and Aerospace Engineering Sciences, University of Colorado, Boulder, CO, 80303, USA
| | - Andy Lin
- Office of Advanced Research Computing, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Zhong Zheng
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Stella Nuo Dong
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Xinli Zhang
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Benjamin M Wu
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Forsyth Institute, Cambridge, MA, 02142, USA.
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Kang Ting
- Forsyth Institute, Cambridge, MA, 02142, USA.
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
6
|
Teng Z, Zhu Y, Lin D, Hao Q, Yue Q, Yu X, Sun S, Jiang L, Lu S. Deciphering the chromatin spatial organization landscapes during BMMSC differentiation. J Genet Genomics 2023; 50:264-275. [PMID: 36720443 DOI: 10.1016/j.jgg.2023.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/18/2023] [Accepted: 01/18/2023] [Indexed: 01/31/2023]
Abstract
The differentiation imbalance in bone marrow mesenchymal stem cells (BMMSCs) is critical for the development of bone density diseases as the population ages. BMMSCs are precursor cells for osteoblasts and adipocytes; however, the chromatin organization landscapes during BMMSC differentiation remain elusive. In this study, we systematically delineate the four-dimensional (4D) genome and dynamic epigenetic atlas of BMMSCs by RNA sequencing (RNA-seq), assay for transposase-accessible chromatin sequencing (ATAC-seq), and high-throughput chromosome conformation capture (Hi-C). The structure analyses reveal 17.5% common and 28.5%-30% specific loops among BMMSCs, osteoblasts, and adipocytes. The subsequent correlation of genome-wide association studies (GWAS) and expression quantitative trait locus (eQTL) data with multi-omics analysis reveal 274 genes and 3634 single nucleotide polymorphisms (SNPs) associated with bone degeneration and osteoporosis (OP). We hypothesize that SNP mutations affect transcription factor (TF) binding sites, thereby affecting changes in gene expression. Furthermore, 26 motifs, 260 TFs, and 291 SNPs are identified to affect the eQTL. Among these genes, DAAM2, TIMP2, and TMEM241 were found to be essential for diseases such as bone degeneration and OP and may serve as potential drug targets.
Collapse
Affiliation(s)
- Zhaowei Teng
- Department of Orthopedics, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, China; Key Laboratory of Yunnan Provincial Innovative Application of Traditional Chinese Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, China; Clinical Medical Research Center, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, China.
| | - Yun Zhu
- The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, Yunnan 653100, China
| | - Da Lin
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Qinggang Hao
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan 650504, China
| | - Qiaoning Yue
- The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, Yunnan 653100, China
| | - Xiaochao Yu
- The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, Yunnan 653100, China
| | - Shuo Sun
- The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, Yunnan 653100, China
| | - Lihong Jiang
- Key Laboratory of Yunnan Provincial Innovative Application of Traditional Chinese Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, China.
| | - Sheng Lu
- Department of Orthopedics, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, China.
| |
Collapse
|
7
|
Griko YV, Loftus DJ, Stolc V, Peletskaya E. Private Spaceflight: A New Landscape for Dealing with Medical Risk. LIFE SCIENCES IN SPACE RESEARCH 2022; 33:41-47. [PMID: 35491028 DOI: 10.1016/j.lssr.2022.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/07/2022] [Accepted: 03/21/2022] [Indexed: 06/14/2023]
Abstract
As NASA and other space agencies make plans to proceed with human exploration missions beyond low earth orbit (LEO), the private sector, including Space X, Virgin Galactic, Blue Origin, Space Adventures and others, echo these plans with initiatives of their own to send humans further into space. Development of more sub-orbital flight opportunities, orbital flight opportunities to LEO and even higher risk endeavors will certainly result in exposure to medical risks for an expanding and heterogeneous population of civilians. To date, a handful of "space tourists" have flown to the International Space Station (ISS), at their own expense, ushering in a new era in which anyone with reasonably good health and even those with physical disability may consider becoming space travelers. Indeed, medical and behavioral issues of healthy, professional astronauts, have not been problematic on short orbital flights. However, recent attempts to test the potential limitations in astronauts on extended duration orbital flights in preparation for future missions beyond LEO raise concern about individual differences in ability to tolerate the hazardous spaceflight environment. Given the rapid development of opportunities for non-professionals and the employees of private companies to travel into space, this is an appropriate time to consider the development of selection strategies for non-government space travelers, including the development of genomic and other modern tools to assess susceptibility to spaceflight risk.
Collapse
Affiliation(s)
- Yuri V Griko
- Division of Space Biosciences, NASA Ames Research Center, Moffett Field, CA, 94035.
| | - David J Loftus
- Division of Space Biosciences, NASA Ames Research Center, Moffett Field, CA, 94035
| | - Viktor Stolc
- Division of Space Biosciences, NASA Ames Research Center, Moffett Field, CA, 94035
| | - Elena Peletskaya
- Analytical Development Department, Bristol-Myers Squibb, Seattle, WA
| |
Collapse
|
8
|
Duan C, Townley HE. Isolation of NELL 1 Aptamers for Rhabdomyosarcoma Targeting. Bioengineering (Basel) 2022; 9:bioengineering9040174. [PMID: 35447734 PMCID: PMC9032205 DOI: 10.3390/bioengineering9040174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/06/2022] [Accepted: 04/13/2022] [Indexed: 12/13/2022] Open
Abstract
NELL1 (Neural epidermal growth factor-like (EGFL)-like protein) is an important biomarker associated with tissue and bone development and regeneration. NELL1 upregulation has been linked with metastasis and negative prognosis in rhabdomyosarcoma (RMS). Furthermore, multiple recent studies have also shown the importance of NELL1 in inflammatory bowel disease and membranous nephropathy, amongst other diseases. In this study, several anti-NELL1 DNA aptamers were selected from a randomized ssDNA pool using a fluorescence-guided method and evaluated for their binding affinity and selectivity. Several other methods such as a metabolic assay and confocal microscopy were also applied for the evaluation of the selected aptamers. The top three candidates were evaluated further, and AptNCan3 was shown to have a binding affinity up to 959.2 nM. Selectivity was examined in the RH30 RMS cells that overexpressed NELL1. Both AptNCan2 and AptNCan3 could significantly suppress metabolic activity in RMS cells. AptNCan3 was found to locate on the cell membrane and also on intracellular vesicles, which matched the location of NELL1 shown by antibodies in previous research. These results indicate that the selected anti-NELL1 aptamer showed strong and highly specific binding to NELL1 and therefore has potential to be used for in vitro or in vivo studies and treatments.
Collapse
Affiliation(s)
- Chengchen Duan
- Nuffield Department of Women’s and Reproductive Health, Oxford University John Radcliffe Hospital, Oxford OX3 9DU, UK;
| | - Helen Elizabeth Townley
- Nuffield Department of Women’s and Reproductive Health, Oxford University John Radcliffe Hospital, Oxford OX3 9DU, UK;
- Department of Engineering Science, Oxford University, Oxford OX1 3PJ, UK
- Correspondence: ; Tel.: +44-1865-283792
| |
Collapse
|
9
|
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: 6] [Impact Index Per Article: 2.0] [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
|
10
|
Bae JH, Park D. Effect of dietary calcium on the gender-specific association between polymorphisms in the PTPRD locus and osteoporosis. Clin Nutr 2022; 41:680-686. [DOI: 10.1016/j.clnu.2022.01.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 01/01/2022] [Accepted: 01/21/2022] [Indexed: 11/03/2022]
|
11
|
Qi H, Kim JK, Ha P, Chen X, Chen E, Chen Y, Li J, Pan HC, Yu M, Mohazeb Y, Azer S, Baik L, Kwak JH, Ting K, Zhang X, Hu M, Soo C. Inactivation of Nell-1 in Chondrocytes Significantly Impedes Appendicular Skeletogenesis. J Bone Miner Res 2019; 34:533-546. [PMID: 30352124 PMCID: PMC6677149 DOI: 10.1002/jbmr.3615] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 09/18/2018] [Accepted: 10/06/2018] [Indexed: 12/29/2022]
Abstract
NELL-1, an osteoinductive protein, has been shown to regulate skeletal ossification. Interestingly, an interstitial 11p14.1-p15.3 deletion involving the Nell-1 gene was recently reported in a patient with short stature and delayed fontanelle closure. Here we sought to define the role of Nell-1 in endochondral ossification by investigating Nell-1-specific inactivation in Col2α1-expressing cell lineages. Nell-1flox/flox ; Col2α1-Cre+ (Nell-1Col2α1 KO) mice were generated for comprehensive analysis. Nell-1Col2α1 KO mice were born alive but displayed subtle femoral length shortening. At 1 and 3 months postpartum, Nell-1 inactivation resulted in dwarfism and premature osteoporotic phenotypes. Specifically, Nell-1Col2α1 KO femurs and tibias exhibited significantly reduced length, bone mineral density (BMD), bone volume per tissue volume (BV/TV), trabecular number/thickness, cortical volume/thickness/density, and increased trabecular separation. The decreased bone formation rate revealed by dynamic histomorphometry was associated with altered numbers and/or function of osteoblasts and osteoclasts. Furthermore, longitudinal observations by in vivo micro-CT showed delayed and reduced mineralization at secondary ossification centers in mutants. Histologically, reduced staining intensities of Safranin O, Col-2, Col-10, and fewer BrdU-positive chondrocytes were observed in thinner Nell-1Col2α1 KO epiphyseal plates along with altered distribution and weaker expression level of Ihh, Patched-1, PTHrP, and PTHrP receptor. Primary Nell-1Col2α1 KO chondrocytes also exhibited decreased proliferation and differentiation, and its downregulated expression of the Ihh-PTHrP signaling molecules can be partially rescued by exogenous Nell-1 protein. Moreover, intranuclear Gli-1 protein and gene expression of the Gli-1 downstream target genes, Hip-1 and N-Myc, were also significantly decreased with Nell-1 inactivation. Notably, the rescue effects were diminished/reduced with application of Ihh signaling inhibitors, cyclopamine or GANT61. Taken together, these findings suggest that Nell-1 is a pivotal modulator of epiphyseal homeostasis and endochondral ossification. The cumulative chondrocyte-specific Nell-1 inactivation significantly impedes appendicular skeletogenesis resulting in dwarfism and premature osteoporosis through inhibiting Ihh signaling and predominantly altering the Ihh-PTHrP feedback loop. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Huichuan Qi
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, Changchun, Jilin, P. R. China
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Jong Kil Kim
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Pin Ha
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Xiaoyan Chen
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
- Department of Orthodontics, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Eric Chen
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Yao Chen
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Jiayi Li
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Hsin Chuan Pan
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Mengliu Yu
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
- Center of Stomatology, China-Japan Friendship Hospital, 2nd Yinghuayuan East Street, Chaoyang District, Beijing, P. R. China
| | - Yasamin Mohazeb
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Sophia Azer
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Lloyd Baik
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Jin Hee Kwak
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Kang Ting
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Xinli Zhang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Min Hu
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, Changchun, Jilin, P. R. China
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
12
|
Appelman-Dijkstra NM, Papapoulos SE. Clinical advantages and disadvantages of anabolic bone therapies targeting the WNT pathway. Nat Rev Endocrinol 2018; 14:605-623. [PMID: 30181608 DOI: 10.1038/s41574-018-0087-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The WNT signalling pathway is a key regulator of bone metabolism, particularly bone formation, which has helped to define the role of osteocytes - the most abundant bone cells - as orchestrators of bone remodelling. Several molecules involved in the control of the WNT signalling pathway have been identified as potential targets for the development of bone-building therapeutics for patients with osteoporosis. Several of these molecules have been investigated in animal models, but only inhibitors of sclerostin (which is produced by osteocytes) have been investigated in phase III clinical studies. Here, we review the rationale for these developments and the specificity and potential off-target actions of WNT-based therapeutics. We also describe the available preclinical and clinical studies and discuss the benefits and risks of using sclerostin inhibitors for the management of patients with osteoporosis.
Collapse
|
13
|
Correa-Rodríguez M, Schmidt Rio-Valle J, Rueda-Medina B. AKAP11 gene polymorphism is associated with bone mass measured by quantitative ultrasound in young adults. Int J Med Sci 2018; 15:999-1004. [PMID: 30013441 PMCID: PMC6036159 DOI: 10.7150/ijms.25369] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 05/27/2018] [Indexed: 01/18/2023] Open
Abstract
Background: Due to the increased prevalence of osteoporosis and direct health care cost of osteoporosis-related fractures, there is a growing interest in identifying genetic markers associated with osteoporosis phenotypes in order to develop genetic screening strategies. We aimed to analyze the possible associations between calcaneal Quantitative ultrasound (QUS), a valuable screening tool for assessing bone status in clinical practice, and ZBTB40 (rs7524102, rs6426749), SP7 (rs2016266) and AKAP11 (rs9533090) genes. Methods: A cross-sectional study was conducted on 550 healthy individuals of Caucasian ancestry (381 females and 169 males, median age 20.46±2,69). Bone mass was assessed through QUS to determine broadband ultrasound attenuation (BUA, dB/MHz). Single-nucleotide polymorphisms (SNPs) in ZBTB40 (rs7524102, rs6426749), SP7 (rs2016266) and AKAP11 (rs9533090) were selected as genetic markers and genotyped using TaqMan OpenArray® technology. Results: Linear regression analysis revealed that rs7524102 and rs6426749 in ZBTB40, and rs9533090 in AKAP11 were significantly associated with the calcaneal QUS parameter after adjustments for age, sex, weight, height, physical activity, and calcium intake (p=0.038, p=0.012 and p=0.008, respectively). After applying the Bonferroni correction for multiple testing (p=0.012), only the association of rs9533090 in AKAP11 remained significant. Conclusion:AKAP11 gene (rs9533090) influences QUS trait in a population of Caucasian young adults. The rs9533090 SNP may be considered a factor affecting peak bone mass acquisition.
Collapse
|
14
|
Rivadeneira F, Uitterlinden AG. Scrutinizing the Genetic Underpinnings of Bone Strength. J Bone Miner Res 2017; 32:2147-2150. [PMID: 29023918 DOI: 10.1002/jbmr.3300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 09/27/2017] [Accepted: 09/27/2017] [Indexed: 11/06/2022]
Affiliation(s)
- Fernando Rivadeneira
- Department of Internal Medicine, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Andre G Uitterlinden
- Department of Internal Medicine, Erasmus University Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
15
|
Karasik D, Demissie S, Lu D, Broe KE, Boyd SK, Liu CT, Hsu YH, Bouxsein ML, Kiel DP. Bone Strength Estimated by Micro-Finite Element Analysis (µFEA) Is Heritable and Shares Genetic Predisposition With Areal BMD: The Framingham Study. J Bone Miner Res 2017; 32:2151-2156. [PMID: 28722129 PMCID: PMC5685872 DOI: 10.1002/jbmr.3200] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 06/06/2017] [Accepted: 06/10/2017] [Indexed: 11/07/2022]
Abstract
Genetic factors contribute to the risk of bone fractures, partly because of effects on bone strength. High-resolution peripheral quantitative computed tomography (HR-pQCT) estimates bone strength using micro-finite element analysis (µFEA). The goal of this study was to investigate if the bone failure load estimated by HR-pQCT-based µFEA is heritable and to what extent it shares genetic regulation with areal bone mineral density (aBMD). Bone microarchitecture was measured by HR-pQCT at the ultradistal tibia and ultradistal radius in adults from the Framingham Heart Study (n = 1087, mean age 72 years; 57% women). Radial and tibial failure load in compression were estimated by µFEA. Femoral neck (FN) and ultradistal forearm (UD) aBMD were measured by dual-energy X-ray absorptiometry (DXA). Heritability (h2 ) of failure load and aBMD and genetic correlations between them was estimated adjusting for covariates (age and sex). Failure load values at the non-weight-bearing ultradistal radius and at the weight-bearing ultradistal tibia were highly correlated (r = 0.906; p < 0.001). Estimates of h2 adjusted for covariates were 0.522 for the radius and 0.497 for the tibia. Additional adjustment for height did not impact on the h2 results, but adjustment for aBMD at the UD and FN somewhat decreased h2 point estimates: 0.222 and 0.380 for radius and tibia, respectively. In bivariate analysis, there was a high phenotypic and genetic correlation between covariate-adjusted failure load at the radius and UD aBMD (ρP = 0.826, ρG = 0.954, respectively), whereas environmental correlations were lower (ρE = 0.696), all highly significant (p < 0.001). Similar correlations were observed between tibial failure load and femoral neck aBMD (ρP = 0.577, ρG = 0.703, both p < 0.001; ρE = 0.432, p < 0.05). These data from adult members of families from a population-based cohort suggest that bone strength of distal extremities estimated by micro-finite element analysis is heritable and shares some genetic composition with areal BMD, regardless of the skeletal site. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- David Karasik
- Institute for Aging Research, Hebrew SeniorLife, Boston, MA
- Faculty of Medicine in the Galilee, Bar Ilan University, Safed, Israel
| | - Serkalem Demissie
- Biostatistics, Boston University School of Public Health, Boston, MA
| | - Darlene Lu
- Biostatistics, Boston University School of Public Health, Boston, MA
| | - Kerry E. Broe
- Institute for Aging Research, Hebrew SeniorLife, Boston, MA
| | - Steven K. Boyd
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Ching-Ti Liu
- Biostatistics, Boston University School of Public Health, Boston, MA
| | - Yi-Hsiang Hsu
- Institute for Aging Research, Hebrew SeniorLife, Boston, MA
- Harvard Medical School, Boston, MA
- Broad Institute of Harvard & MIT
| | - Mary L. Bouxsein
- Center for Advanced Orthopedic Studies, Beth Israel Deaconess Medical Center
- Harvard Medical School, Boston, MA
| | - Douglas P. Kiel
- Institute for Aging Research, Hebrew SeniorLife, Boston, MA
- Harvard Medical School, Boston, MA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
- Broad Institute of Harvard & MIT
| |
Collapse
|
16
|
Correa-Rodríguez M, Schmidt-RioValle J, Rueda-Medina B. The rs3736228 polymorphism in the LRP5 gene is associated with calcaneal ultrasound parameter but not with body composition in a cohort of young Caucasian adults. J Bone Miner Metab 2017; 35:694-700. [PMID: 28028632 DOI: 10.1007/s00774-016-0808-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 11/15/2016] [Indexed: 01/27/2023]
Abstract
The aim of the present study was to investigate the possible influence of low-density lipoprotein receptor-related protein 5 (LRP5) and sclerostin (SOST) genes as genetic factors contributing to calcaneal quantitative ultrasound (QUS) and body composition variables in a population of young Caucasian adults. The study population comprised a total of 575 individuals (mean age 20.41years; SD 2.36) whose bone mass was assessed through QUS to determine broadband ultrasound attenuation (BUA, dB/MHz). Body composition measurements were performed using a body composition analyser. Seven single-nucleotide polymorphisms (SNPs) of LRP5 (rs2306862, rs599083, rs556442 and rs3736228) and SOST (rs4792909, rs851054 and rs2023794) were selected as genetic markers and genotyped using TaqMan OpenArray® technology. Linear regression analysis was used to test the possible association of the tested SNPs with QUS and body composition parameters. Linear regression analysis revealed that the rs3736228 SNP of LPR5 was significantly associated with BUA after adjustment for age, sex, weight, height, physical activity and calcium intake (P = 0.028, β (95% CI) = 0.089 (0.099-1.691). For the remaining SNPs, no significant association with the QUS measurement was observed. Regarding body composition, no significant association was found between LRP5 and SOST polymorphisms and body mass index, total fat mass and total lean mass after adjustment for age and sex as covariates. We concluded that the rs3736228 LRP5 genetic polymorphism influences calcaneal QUS parameter in a population of young Caucasian adults. This finding suggests that LRP5 might be an important genetic marker contributing to bone mass accrual early in life.
Collapse
Affiliation(s)
- María Correa-Rodríguez
- Faculty of Health Sciences, University of Granada, Av. Ilustración S/N, 18007, Granada, Spain.
| | | | - Blanca Rueda-Medina
- Faculty of Health Sciences, University of Granada, Av. Ilustración S/N, 18007, Granada, Spain
| |
Collapse
|
17
|
Pakvasa M, Alverdy A, Mostafa S, Wang E, Fu L, Li A, Oliveira L, Athiviraham A, Lee MJ, Wolf JM, He TC, Ameer GA, Reid RR. Neural EGF-like protein 1 (NELL-1): Signaling crosstalk in mesenchymal stem cells and applications in regenerative medicine. Genes Dis 2017; 4:127-137. [PMID: 29276737 PMCID: PMC5737940 DOI: 10.1016/j.gendis.2017.07.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 07/27/2017] [Indexed: 12/15/2022] Open
Abstract
Bone tissue regeneration holds the potential to solve both osteoporosis and large skeletal defects, two problems associated with significant morbidity. The differentiation of mesenchymal stem cells into the osteogenic lineage requires a specific microenvironment and certain osteogenic growth factors. Neural EGF Like-Like molecule 1 (NELL-1) is a secreted glycoprotein that has proven, both in vitro and in vivo, to be a potent osteo-inductive factor. Furthermore, it has been shown to repress adipogenic differentiation and inflammation. NELL-1 can work synergistically with other osteogenic factors such as Bone Morphogenic Protein (BMP) -2 and -9, and has shown promise for use in tissue engineering and as a systemically administered drug for the treatment of osteoporosis. Here we provide a comprehensive up-to-date review on the molecular signaling cascade of NELL-1 in mesenchymal stem cells and potential applications in bone regenerative engineering.
Collapse
Affiliation(s)
- Mikhail Pakvasa
- The University of Chicago, Pritzker School of Medicine, Chicago, IL 60637, USA
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Alex Alverdy
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Rosalind Franklin University, Chicago Medical School, North Chicago, IL 60064, USA
| | - Sami Mostafa
- The University of Chicago, Pritzker School of Medicine, Chicago, IL 60637, USA
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Eric Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Lucy Fu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Alexander Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Leonardo Oliveira
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Aravind Athiviraham
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael J. Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jennifer Moriatis Wolf
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Guillermo A. Ameer
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Russell R. Reid
- The University of Chicago, Pritzker School of Medicine, Chicago, IL 60637, USA
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| |
Collapse
|
18
|
Abstract
BACKGROUND The receptor activator of the nuclear factor-kappa B ligand (RANKL), the receptor activator of nuclear factor-kappa B (RANK), and the osteoprotegerin (OPG) signaling pathway play an important role in the regulation of bone remodeling and osteoclast differentiation. Quantitative ultrasound (QUS) is a relatively recent and noninvasive method providing structural information on microstructure, bone elasticity, and connectivity. However, in contrast to bone mineral density measurements, the possible association of the RANKL/RANK/OPG pathway with heel QUS has not been analyzed. OBJECTIVES The aim of this study was to assess, for the first time, the contribution of the RANKL/RANK/OPG pathway genes in the genetic background of heel QUS parameters. METHODS Ten single-nucleotide polymorphisms (SNPs) of RANKL (rs9594759, rs12585014, rs7988338, rs2148073), RANK (rs1805034, rs12458117, rs3018362), and OPG (rs4355801, rs3102735, rs2073618) were selected as genetic markers and genotyped using Open Array technology in 575 self-reported Caucasian individuals aged 18-25. Bone mass in the right calcaneus was estimated with QUS to obtain the broadband ultrasound attenuation (BUA) measurement (dB/MHz). Linear regression analyses were performed to test the possible association between the SNPs and BUA. RESULTS Linear regression analysis of all the tested SNPs revealed no significant association with the BUA parameter after adjusting for age, gender, weight, height, physical activity, and calcium intake. The lowest p-value was observed for the rs9594759 RANKL polymorphism and heel QUS (p = .06; b* = -.075, 95% CI [-0.960, 0.028]). CONCLUSION Our results suggest that the polymorphism of the RANKL, RANK, and OPG genes does not make a significant genetic contribution to heel ultrasound measurements in a population of young Caucasian adults. Further studies replicating the results in independent populations are needed to support these initial findings.
Collapse
|
19
|
Karasik D, Demissie S, Zhou Y, Lu D, Broe KE, Bouxsein ML, Cupples LA, Kiel DP. Heritability and Genetic Correlations for Bone Microarchitecture: The Framingham Study Families. J Bone Miner Res 2017; 32:106-114. [PMID: 27419666 PMCID: PMC5310688 DOI: 10.1002/jbmr.2915] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 06/13/2016] [Accepted: 07/11/2016] [Indexed: 02/02/2023]
Abstract
High-resolution peripheral quantitative computed tomography (HR-pQCT) measures bone microarchitecture and volumetric bone mineral density (vBMD), important risk factors for osteoporotic fractures. We estimated the heritability (h2 ) of bone microstructure indices and vBMD, measured by HR-pQCT, and genetic correlations (ρG ) among them and between them and regional aBMD measured by dual-energy X-ray absorptiometry (DXA), in adult relatives from the Framingham Heart Study. Cortical (Ct) and trabecular (Tb) traits were measured at the distal radius and tibia in up to 1047 participants, and ultradistal radius (UD) aBMD was obtained by DXA. Heritability estimates, adjusted for age, sex, and estrogenic status (in women), ranged from 19.3% (trabecular number) to 82.8% (p < 0.01, Ct.vBMD) in the radius and from 51.9% (trabecular thickness) to 98.3% (cortical cross-sectional area fraction) in the tibia. Additional adjustments for height, weight, and radial aBMD had no major effect on h2 estimates. In bivariate analyses, moderate to high genetic correlations were found between radial total vBMD and microarchitecture traits (ρG from 0.227 to 0.913), except for cortical porosity. At the tibia, a similar pattern of genetic correlations was observed (ρG from 0.274 to 0.948), except for cortical porosity. Environmental correlations between the microarchitecture traits were also substantial. There were high genetic correlations between UD aBMD and multivariable-adjusted total and trabecular vBMD at the radius (ρG = 0.811 and 0.917, respectively). In summary, in related men and women from a population-based cohort, cortical and trabecular microarchitecture and vBMD at the radius and tibia were heritable and shared some h2 with regional aBMD measured by DXA. These findings of high heritability of HR-pQCT traits, with a slight attenuation when adjusting for aBMD, supports further work to identify the specific variants underlying volumetric bone density and fine structure of long bones. Knowledge that some of these traits are genetically correlated can serve to reduce the number of traits for genetic association studies. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- David Karasik
- Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA.,Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Serkalem Demissie
- Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Yanhua Zhou
- Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Darlene Lu
- Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Kerry E Broe
- Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA
| | - Mary L Bouxsein
- Center for Advanced Orthopedic Studies, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - L Adrienne Cupples
- Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA.,Framingham Heart Study, Framingham, MA, USA
| | - Douglas P Kiel
- Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
20
|
Tanjaya J, Zhang Y, Lee S, Shi J, Chen E, Ang P, Zhang X, Tetradis S, Ting K, Wu B, Soo C, Kwak JH. Efficacy of Intraperitoneal Administration of PEGylated NELL-1 for Bone Formation. Biores Open Access 2016; 5:159-70. [PMID: 27354930 PMCID: PMC4921932 DOI: 10.1089/biores.2016.0018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Systemically delivered NEL-like molecule-1 (NELL-1), a potent pro-osteogenic protein, promotes bone formation in healthy and osteoporotic mouse models. PEGylation of NELL-1 (NELL-PEG) increases the half-life of the protein in a mouse model without compromising its osteogenic potential, thereby improving its pharmacokinetics upon systemic delivery. This study consists of a twofold approach: a biodistribution test and an in vivo osteogenic potential test. The biodistribution test compared two commonly used administration methods for drug delivery other than intravenous-intraperitoneal (IP) and subcutaneous (SC)-to examine NELL-PEG biodistribution in mice. Compared to a single-dose SC injection (1.25 mg/kg), a single-dose IP administration yielded a higher protein uptake in the targeted bone sites. When the IP injection dose was doubled to 2.5 mg/kg, the protein remained in the femurs, tibias, and vertebrae for up to 72 h. Next, based on the results of the biodistribution study, IP administration was selected to further investigate the in vivo osteogenic effects of weekly NELL-PEG injection (q7d). In vivo, the IP administered NELL-PEG group showed significantly greater bone mineral density, bone volume fraction, and trabecular bone formation in the targeted bone sites compared to the phosphate-buffered saline control. In summary, weekly NELL-PEG injection via IP administration successfully enhanced the overall bone quality. These findings demonstrate that systemic delivery of NELL-PEG via IP administration may serve as an effective osteogenic therapy for preventing and treating osteoporosis.
Collapse
Affiliation(s)
- Justine Tanjaya
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Yulong Zhang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California.; Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, California
| | - Soonchul Lee
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University, Pocheon, South Korea.; Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Jiayu Shi
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Eric Chen
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Pia Ang
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California.; Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Xinli Zhang
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Sotirios Tetradis
- Section of Oral and Maxillofacial Radiology, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Kang Ting
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California.; Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Benjamin Wu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California.; Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, California
| | - Chia Soo
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California.; Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Jin Hee Kwak
- Division of Growth and Development and the Section of Orthodontics, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| |
Collapse
|
21
|
James AW, Chiang M, Asatrian G, Shen J, Goyal R, Chung CG, Chang L, Shrestha S, Turner AS, Seim HB, Zhang X, Wu BM, Ting K, Soo C. Vertebral Implantation of NELL-1 Enhances Bone Formation in an Osteoporotic Sheep Model. Tissue Eng Part A 2016; 22:840-9. [PMID: 27113550 DOI: 10.1089/ten.tea.2015.0230] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Vertebral compression fractures related to osteoporosis greatly afflict the aging population. One of the most commonly used therapy today is balloon kyphoplasty. However, this treatment is far from ideal and is associated with significant side effects. NELL-1, an osteoinductive factor that possesses both pro-osteogenic and anti-osteoclastic properties, is a promising candidate for an alternative to current treatment modalities. This study utilizes the pro-osteogenic properties of recombinant human NELL-1 (rhNELL-1) in lumbar spine vertebral defect model in osteoporotic sheep. METHODS Osteoporosis was induced through ovariectomy, dietary depletion of calcium and vitamin D, and steroid administration. After osteoporotic induction, lumbar vertebral body defect creation was performed. Sheep were randomly implanted with the control vehicle, comprised of hyaluronic acid (HA) with hydroxyapatite-coated β-tricalcium phosphate (β-TCP), or the treatment material of rhNELL-1 protein lyophilized onto β-TCP mixed with HA. Analysis of lumbar spine defect healing was performed by radiographic, histologic, and computer-simulated biomechanical testing. RESULTS rhNELL-1 treatment significantly increased lumbar spine bone formation, as determined by bone mineral density, % bone volume, and mean cortical width as assessed by micro-computed tomography. Histological analysis revealed a significant increase in bone area and osteoblast number and decrease in osteoclast number around the implant site. Computer-simulated biomechanical analysis of trabecular bone demonstrated that rhNELL-1-treatment resulted in a significantly more stress-resistant composition. CONCLUSION Our findings suggest rhNELL-1-based vertebral implantation successfully improved cortical and cancellous bone regeneration in the lumbar spine of osteoporotic sheep. rhNELL-1-based bone graft substitutes represent a potential new local therapy.
Collapse
Affiliation(s)
- Aaron W James
- 1 Departments of Surgery and Orthopaedic Surgery, Orthopaedic Hospital Research Center, UCLA and Orthopedic Hospital , Los Angeles, California.,2 Division of Growth and Development and Section of Orthodontics, Dental and Craniofacial Research Institute, School of Dentistry, University of California , Los Angeles, Los Angeles, California.,3 Department of Pathology and Laboratory Medicine, University of California , Los Angeles, Los Angeles, California
| | - Michael Chiang
- 2 Division of Growth and Development and Section of Orthodontics, Dental and Craniofacial Research Institute, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Greg Asatrian
- 2 Division of Growth and Development and Section of Orthodontics, Dental and Craniofacial Research Institute, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Jia Shen
- 1 Departments of Surgery and Orthopaedic Surgery, Orthopaedic Hospital Research Center, UCLA and Orthopedic Hospital , Los Angeles, California.,2 Division of Growth and Development and Section of Orthodontics, Dental and Craniofacial Research Institute, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Raghav Goyal
- 2 Division of Growth and Development and Section of Orthodontics, Dental and Craniofacial Research Institute, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Choon G Chung
- 2 Division of Growth and Development and Section of Orthodontics, Dental and Craniofacial Research Institute, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Le Chang
- 1 Departments of Surgery and Orthopaedic Surgery, Orthopaedic Hospital Research Center, UCLA and Orthopedic Hospital , Los Angeles, California.,2 Division of Growth and Development and Section of Orthodontics, Dental and Craniofacial Research Institute, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Swati Shrestha
- 1 Departments of Surgery and Orthopaedic Surgery, Orthopaedic Hospital Research Center, UCLA and Orthopedic Hospital , Los Angeles, California.,2 Division of Growth and Development and Section of Orthodontics, Dental and Craniofacial Research Institute, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - A Simon Turner
- 4 Department of Veterinary Sciences, Colorado State University , Fort Collins, Colorado
| | - Howard B Seim
- 4 Department of Veterinary Sciences, Colorado State University , Fort Collins, Colorado
| | - Xinli Zhang
- 1 Departments of Surgery and Orthopaedic Surgery, Orthopaedic Hospital Research Center, UCLA and Orthopedic Hospital , Los Angeles, California.,2 Division of Growth and Development and Section of Orthodontics, Dental and Craniofacial Research Institute, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Benjamin M Wu
- 5 Departments of Bioengineering and Material Sciences, University of California , Los Angeles, Los Angeles, California
| | - Kang Ting
- 1 Departments of Surgery and Orthopaedic Surgery, Orthopaedic Hospital Research Center, UCLA and Orthopedic Hospital , Los Angeles, California.,2 Division of Growth and Development and Section of Orthodontics, Dental and Craniofacial Research Institute, School of Dentistry, University of California , Los Angeles, Los Angeles, California
| | - Chia Soo
- 1 Departments of Surgery and Orthopaedic Surgery, Orthopaedic Hospital Research Center, UCLA and Orthopedic Hospital , Los Angeles, California.,6 Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California , Los Angeles, Los Angeles, California
| |
Collapse
|
22
|
Abstract
Osteoporosis is characterized by low bone mass and an increased risk of fracture. Genetic factors, environmental factors and gene-environment interactions all contribute to a person's lifetime risk of developing an osteoporotic fracture. This Review summarizes key advances in understanding of the genetics of bone traits and their role in osteoporosis. Candidate-gene approaches dominated this field 20 years ago, but clinical and preclinical genetic studies published in the past 5 years generally utilize more-sophisticated and better-powered genome-wide association studies (GWAS). High-throughput DNA sequencing, large genomic databases and improved methods of data analysis have greatly accelerated the gene-discovery process. Linkage analyses of single-gene traits that segregate in families with extreme phenotypes have led to the elucidation of critical pathways controlling bone mass. For example, components of the Wnt-β-catenin signalling pathway have been validated (in both GWAS and functional studies) as contributing to various bone phenotypes. These notable advances in gene discovery suggest that the next decade will witness cataloguing of the hundreds of genes that influence bone mass and osteoporosis, which in turn will provide a roadmap for the development of new drugs that target diseases of low bone mass, including osteoporosis.
Collapse
|
23
|
James AW, Shen J, Zhang X, Asatrian G, Goyal R, Kwak JH, Jiang L, Bengs B, Culiat CT, Turner AS, Seim Iii HB, Wu BM, Lyons K, Adams JS, Ting K, Soo C. NELL-1 in the treatment of osteoporotic bone loss. Nat Commun 2015; 6:7362. [PMID: 26082355 PMCID: PMC4557288 DOI: 10.1038/ncomms8362] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 04/28/2015] [Indexed: 01/09/2023] Open
Abstract
NELL-1 is a secreted, osteoinductive protein whose expression rheostatically controls skeletal ossification. Overexpression of NELL-1 results in craniosynostosis in humans and mice, whereas lack of Nell-1 expression is associated with skeletal undermineralization. Here we show that Nell-1-haploinsufficient mice have normal skeletal development but undergo age-related osteoporosis, characterized by a reduction in osteoblast:osteoclast (OB:OC) ratio and increased bone fragility. Recombinant NELL-1 binds to integrin β1 and consequently induces Wnt/β-catenin signalling, associated with increased OB differentiation and inhibition of OC-directed bone resorption. Systemic delivery of NELL-1 to mice with gonadectomy-induced osteoporosis results in improved bone mineral density. When extended to a large animal model, local delivery of NELL-1 to osteoporotic sheep spine leads to significant increase in bone formation. Altogether, these findings suggest that NELL-1 deficiency plays a role in osteoporosis and demonstrate the potential utility of NELL-1 as a combination anabolic/antiosteoclastic therapeutic for bone loss.
Collapse
Affiliation(s)
- Aaron W James
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA and Orthopaedic Hospital, University of California, Los Angeles, California 90095, USA.,Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California 90095, USA.,Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA
| | - Jia Shen
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA and Orthopaedic Hospital, University of California, Los Angeles, California 90095, USA.,Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California 90095, USA
| | - Xinli Zhang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California 90095, USA
| | - Greg Asatrian
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California 90095, USA
| | - Raghav Goyal
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California 90095, USA
| | - Jin H Kwak
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California 90095, USA
| | - Lin Jiang
- Department of Neurology, Easton Center for Alzheimer's Disease Research, Molecular Biology Institute, University of California, Los Angeles, California 90095, USA
| | - Benjamin Bengs
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA and Orthopaedic Hospital, University of California, Los Angeles, California 90095, USA
| | | | - A Simon Turner
- Department of Veterinary Sciences, Colorado State University, Fort Collins, Colorado 80523, USA
| | - Howard B Seim Iii
- Department of Veterinary Sciences, Colorado State University, Fort Collins, Colorado 80523, USA
| | - Benjamin M Wu
- Department of Bioengineering and Department of Material Sciences, University of California, Los Angeles, California 90095, USA
| | - Karen Lyons
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA and Orthopaedic Hospital, University of California, Los Angeles, California 90095, USA
| | - John S Adams
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA and Orthopaedic Hospital, University of California, Los Angeles, California 90095, USA
| | - Kang Ting
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA and Orthopaedic Hospital, University of California, Los Angeles, California 90095, USA.,Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, California 90095, USA
| | - Chia Soo
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA and Orthopaedic Hospital, University of California, Los Angeles, California 90095, USA.,Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
24
|
Johnsson M, Jonsson KB, Andersson L, Jensen P, Wright D. Genetic regulation of bone metabolism in the chicken: similarities and differences to Mammalian systems. PLoS Genet 2015; 11:e1005250. [PMID: 26023928 PMCID: PMC4449198 DOI: 10.1371/journal.pgen.1005250] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 04/28/2015] [Indexed: 11/19/2022] Open
Abstract
Birds have a unique bone physiology, due to the demands placed on them through egg production. In particular their medullary bone serves as a source of calcium for eggshell production during lay and undergoes continuous and rapid remodelling. We take advantage of the fact that bone traits have diverged massively during chicken domestication to map the genetic basis of bone metabolism in the chicken. We performed a quantitative trait locus (QTL) and expression QTL (eQTL) mapping study in an advanced intercross based on Red Junglefowl (the wild progenitor of the modern domestic chicken) and White Leghorn chickens. We measured femoral bone traits in 456 chickens by peripheral computerised tomography and femoral gene expression in a subset of 125 females from the cross with microarrays. This resulted in 25 loci for female bone traits, 26 loci for male bone traits and 6318 local eQTL loci. We then overlapped bone and gene expression loci, before checking for an association between gene expression and trait values to identify candidate quantitative trait genes for bone traits. A handful of our candidates have been previously associated with bone traits in mice, but our results also implicate unexpected and largely unknown genes in bone metabolism. In summary, by utilising the unique bone metabolism of an avian species, we have identified a number of candidate genes affecting bone allocation and metabolism. These findings can have ramifications not only for the understanding of bone metabolism genetics in general, but could also be used as a potential model for osteoporosis as well as revealing new aspects of vertebrate bone regulation or features that distinguish avian and mammalian bone. In this work we seek to further the understanding of bone genetics by mapping bone traits and gene expression in the chicken. Bone in female birds is special due to egg production. In this study, we combine the genetic mapping of bone traits with bone gene expression to find candidate quantitative trait genes that explain the differences between wild and domestic chickens in terms of bone production. The concept of combining genetic mapping and gene expression mapping is not new, and has already been successful in isolating bone-related genes in mammals, however this is the first time it has been applied to an avian system with such unique bone modelling processes. We aim to reveal new molecular mechanisms of bone regulation, and many of the candidates we find are new, highlighting the potential this technique has to identify the potential differences between avian and mammalian bone biology.
Collapse
Affiliation(s)
- Martin Johnsson
- AVIAN Behavioural Genomics and Physiology group, IFM Biology, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Kenneth B. Jonsson
- Department of Surgical Sciences, Orthopaedics, Akademiska Sjukhuset, Uppsala University, Uppsala, Sweden
| | - Leif Andersson
- Department of Medical Biochemistry and Microbiology, BMC, Uppsala University, Uppsala, Sweden
| | - Per Jensen
- AVIAN Behavioural Genomics and Physiology group, IFM Biology, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Dominic Wright
- AVIAN Behavioural Genomics and Physiology group, IFM Biology, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
- * E-mail:
| |
Collapse
|
25
|
Pharmacokinetics and osteogenic potential of PEGylated NELL-1 in vivo after systemic administration. Biomaterials 2015; 57:73-83. [PMID: 25913252 DOI: 10.1016/j.biomaterials.2015.03.063] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 03/28/2015] [Accepted: 03/31/2015] [Indexed: 12/11/2022]
Abstract
Osteoporosis is a skeletal disorder attributable to an imbalance in osteoblast and osteoclast activity. NELL-1, a secretory protein that promotes osteogenesis while suppressing osteoclastic activity, holds potential as an osteoporosis therapy. Recently, we demonstrated that PEGylation of NELL-1 significantly improves its thermostability while preserving its bioactivity in vitro. However, the effect of PEGylation on the pharmacokinetics and osteogenic potential of NELL-1 in vivo have yet to be investigated. The present study demonstrated that PEGylation of NELL-1 significantly increases the elimination half-life time of the protein from 5.5 h to 15.5 h while distributing more than 2-3 times the amount of protein to bone tissues (femur, tibia, vertebrae, calvaria) in vivo when compared to naked NELL-1. In addition, microCT and DXA analyses demonstrated that systemic NELL-PEG therapy administered every 4 or 7 days significantly increases not only femoral and lumbar BMD and percent bone volume, but also new bone formation throughout the overall skeleton after four weeks of treatment. Furthermore, immunohistochemistry revealed increased osteocalcin expression, while TRAP staining showed reduced osteoclast numbers in NELL-PEG groups. Our findings suggest that the PEGylation technique presents a viable and promising approach to further develop NELL-1 into an effective systemic therapeutic for the treatment of osteoporosis.
Collapse
|
26
|
Zhang Y, Velasco O, Zhang X, Ting K, Soo C, Wu BM. Bioactivity and circulation time of PEGylated NELL-1 in mice and the potential for osteoporosis therapy. Biomaterials 2014; 35:6614-21. [PMID: 24818884 PMCID: PMC4077898 DOI: 10.1016/j.biomaterials.2014.04.061] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 04/16/2014] [Indexed: 12/12/2022]
Abstract
Osteoporosis is a progressive bone disease due to low osteoblast activity and/or high osteoclast activity. NELL-1 is a potential therapy for osteoporosis because it specifically increases osteoblast differentiation. However, similar to other protein drugs, the bioavailability of NELL-1 may be limited by its in vivo half-life and rapid clearance from body. The purpose of the present study is to prolong NELL-1 circulation time in vivo by PEGylation with three monomeric PEG sizes (5, 20, 40 kDa). While linear PEG 5k yielded the most efficient PEGylation and the most thermally stable conjugate, linear PEG 20k resulted in the conjugate with the highest Mw and longest in vivo circulation. Compared to non-modified NELL-1, all three PEGylated conjugates showed enhanced thermal stability and each prolonged the in vivo circulation time significantly. Furthermore, PEGylated NELL-1 retained its osteoblastic activity without any appreciable cytotoxicity. These findings motivate further studies to evaluate the efficacy of PEGylated NELL-1 on the prevention and treatment of osteoporosis.
Collapse
Affiliation(s)
- Yulong Zhang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Materials Science and Engineering, and Division of Advanced Prosthodontics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Omar Velasco
- Weintraub Center for Reconstructive Biotechnology, and Dental and Craniofacial Research Institute, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xinli Zhang
- Weintraub Center for Reconstructive Biotechnology, and Dental and Craniofacial Research Institute, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kang Ting
- Weintraub Center for Reconstructive Biotechnology, and Dental and Craniofacial Research Institute, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chia Soo
- Weintraub Center for Reconstructive Biotechnology, and Dental and Craniofacial Research Institute, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Benjamin M Wu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Materials Science and Engineering, and Division of Advanced Prosthodontics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Weintraub Center for Reconstructive Biotechnology, and Dental and Craniofacial Research Institute, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
27
|
Hernandez-de Sosa N, Athanasiadis G, Malouf J, Laiz A, Marin A, Herrera S, Farrerons J, Soria JM, Casademont J. Heritability of bone mineral density in a multivariate family-based study. Calcif Tissue Int 2014; 94:590-596. [PMID: 24687525 DOI: 10.1007/s00223-014-9852-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 03/14/2014] [Indexed: 01/01/2023]
Abstract
There is evidence for a genetic contribution to bone mineral density (BMD×). Different loci affecting BMD have been identified by diverse linkage and genome-wide association studies. We studied the heritability of and the correlations among six densitometric phenotypes and four bone mass/fracture phenotypes. For this purpose, we used a family-based study of the genetics of osteoporosis, the Genetic Analysis of Osteoporosis Project. The primary aim of our study was to examine the roles of genetic and environmental factors in determining osteoporosis-related phenotypes. The project consisted of 11 extended families from Spain. All of them were selected through a proband with osteoporosis. BMD was measured using dual-energy X-ray absorptiometry. The proportion of variance of BMD attributable to significant covariates ranged from 25% (for femoral neck BMD) to 48% (for whole-body total BMD). The vast majority of the densitometric phenotypes had highly significant heritability, ranging from 0.252 (whole-body total BMD) to 0.537 (trochanteric BMD) after correcting for covariate effects. All of the densitometric phenotypes showed high and significant genetic correlations (from -0.772 to -1.000) with a low bone mass/osteopenia condition (Affected 3). Our findings provide additional evidence on the heritability of BMD and a strong genetic correlation between BMD and bone mass/fracture phenotypes in a Spanish population. Our results emphasize the importance of detecting genetic risk factors and the benefit of early diagnosis and especially therapeutic and preventive strategies.
Collapse
Affiliation(s)
- Nerea Hernandez-de Sosa
- Department of Internal Medicine, Hospital de la Santa Creu i Sant Pau, Autonomous University of Barcelona, Barcelona, Spain,
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
The musculoskeletal system is a complex organ comprised of the skeletal bones, skeletal muscles, tendons, ligaments, cartilage, joints, and other connective tissue that physically and mechanically interact to provide animals and humans with the essential ability of locomotion. This mechanical interaction is undoubtedly essential for much of the diverse shape and forms observed in vertebrates and even in invertebrates with rudimentary musculoskeletal systems such as fish. It makes sense from a historical point of view that the mechanical theories of musculoskeletal development have had tremendous influence of our understanding of biology, because these relationships are clear and palpable. Less visible to the naked eye or even to the microscope is the biochemical interaction among the individual players of the musculoskeletal system. It was only in recent years that we have begun to appreciate that beyond this mechanical coupling of muscle and bones, these 2 tissues function at a higher level through crosstalk signaling mechanisms that are important for the function of the concomitant tissue. Our brief review attempts to present some of the key concepts of these new concepts and is outline to present muscles and bones as secretory/endocrine organs, the evidence for mutual genetic and tissue interactions, pathophysiological examples of crosstalk, and the exciting new directions for this promising field of research aimed at understanding the biochemical/molecular coupling of these 2 intimately associated tissues.
Collapse
Affiliation(s)
- Marco Brotto
- Muscle Biology Research Group-MUBIG, UMKC School of Nursing & Health Studies and School of Medicine, 2464 Charlotte Street, Kansas City, MO, 64108, USA,
| | | |
Collapse
|
29
|
Bolormaa S, Pryce JE, Reverter A, Zhang Y, Barendse W, Kemper K, Tier B, Savin K, Hayes BJ, Goddard ME. A multi-trait, meta-analysis for detecting pleiotropic polymorphisms for stature, fatness and reproduction in beef cattle. PLoS Genet 2014; 10:e1004198. [PMID: 24675618 PMCID: PMC3967938 DOI: 10.1371/journal.pgen.1004198] [Citation(s) in RCA: 178] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 01/02/2014] [Indexed: 12/14/2022] Open
Abstract
Polymorphisms that affect complex traits or quantitative trait loci (QTL) often affect multiple traits. We describe two novel methods (1) for finding single nucleotide polymorphisms (SNPs) significantly associated with one or more traits using a multi-trait, meta-analysis, and (2) for distinguishing between a single pleiotropic QTL and multiple linked QTL. The meta-analysis uses the effect of each SNP on each of n traits, estimated in single trait genome wide association studies (GWAS). These effects are expressed as a vector of signed t-values (t) and the error covariance matrix of these t values is approximated by the correlation matrix of t-values among the traits calculated across the SNP (V). Consequently, t'V−1t is approximately distributed as a chi-squared with n degrees of freedom. An attractive feature of the meta-analysis is that it uses estimated effects of SNPs from single trait GWAS, so it can be applied to published data where individual records are not available. We demonstrate that the multi-trait method can be used to increase the power (numbers of SNPs validated in an independent population) of GWAS in a beef cattle data set including 10,191 animals genotyped for 729,068 SNPs with 32 traits recorded, including growth and reproduction traits. We can distinguish between a single pleiotropic QTL and multiple linked QTL because multiple SNPs tagging the same QTL show the same pattern of effects across traits. We confirm this finding by demonstrating that when one SNP is included in the statistical model the other SNPs have a non-significant effect. In the beef cattle data set, cluster analysis yielded four groups of QTL with similar patterns of effects across traits within a group. A linear index was used to validate SNPs having effects on multiple traits and to identify additional SNPs belonging to these four groups. We describe novel methods for finding significant associations between a genome wide panel of SNPs and multiple complex traits, and further for distinguishing between genes with effects on multiple traits and multiple linked genes affecting different traits. The method uses a meta-analysis based on estimates of SNP effects from independent single trait genome wide association studies (GWAS). The method could therefore be widely used to combine already published GWAS results. The method was applied to 32 traits that describe growth, body composition, feed intake and reproduction in 10,191 beef cattle genotyped for approximately 700,000 SNP. The genes found to be associated with these traits can be arranged into 4 groups that differ in their pattern of effects and hence presumably in their physiological mechanism of action. For instance, one group of genes affects weight and fatness in the opposite direction and can be described as a group of genes affecting mature size, while another group affects weight and fatness in the same direction.
Collapse
Affiliation(s)
- Sunduimijid Bolormaa
- Victorian Department of Environment and Primary Industries, Bundoora, Victoria, Australia
- * E-mail:
| | - Jennie E. Pryce
- Victorian Department of Environment and Primary Industries, Bundoora, Victoria, Australia
| | - Antonio Reverter
- CSIRO Animal, Food and Health Sciences, Queensland Bioscience Precinct, St. Lucia, Queensland, Australia
| | - Yuandan Zhang
- Animal Genetics and Breeding Unit, University of New England, Armidale, New South Wales, Australia
| | - William Barendse
- CSIRO Animal, Food and Health Sciences, Queensland Bioscience Precinct, St. Lucia, Queensland, Australia
| | - Kathryn Kemper
- School of Land and Environment, University of Melbourne, Parkville, Victoria, Australia
| | - Bruce Tier
- Animal Genetics and Breeding Unit, University of New England, Armidale, New South Wales, Australia
| | - Keith Savin
- Victorian Department of Environment and Primary Industries, Bundoora, Victoria, Australia
| | - Ben J. Hayes
- Victorian Department of Environment and Primary Industries, Bundoora, Victoria, Australia
| | - Michael E. Goddard
- Victorian Department of Environment and Primary Industries, Bundoora, Victoria, Australia
- School of Land and Environment, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
30
|
Moayyeri A, Hsu YH, Karasik D, Estrada K, Xiao SM, Nielson C, Srikanth P, Giroux S, Wilson SG, Zheng HF, Smith AV, Pye SR, Leo PJ, Teumer A, Hwang JY, Ohlsson C, McGuigan F, Minster RL, Hayward C, Olmos JM, Lyytikäinen LP, Lewis JR, Swart KMA, Masi L, Oldmeadow C, Holliday EG, Cheng S, van Schoor NM, Harvey NC, Kruk M, del Greco M F, Igl W, Trummer O, Grigoriou E, Luben R, Liu CT, Zhou Y, Oei L, Medina-Gomez C, Zmuda J, Tranah G, Brown SJ, Williams FM, Soranzo N, Jakobsdottir J, Siggeirsdottir K, Holliday KL, Hannemann A, Go MJ, Garcia M, Polasek O, Laaksonen M, Zhu K, Enneman AW, McEvoy M, Peel R, Sham PC, Jaworski M, Johansson Å, Hicks AA, Pludowski P, Scott R, Dhonukshe-Rutten RAM, van der Velde N, Kähönen M, Viikari JS, Sievänen H, Raitakari OT, González-Macías J, Hernández JL, Mellström D, Ljunggren O, Cho YS, Völker U, Nauck M, Homuth G, Völzke H, Haring R, Brown MA, McCloskey E, Nicholson GC, Eastell R, Eisman JA, Jones G, Reid IR, Dennison EM, Wark J, Boonen S, Vanderschueren D, Wu FCW, Aspelund T, Richards JB, Bauer D, Hofman A, Khaw KT, Dedoussis G, Obermayer-Pietsch B, Gyllensten U, Pramstaller PP, Lorenc RS, et alMoayyeri A, Hsu YH, Karasik D, Estrada K, Xiao SM, Nielson C, Srikanth P, Giroux S, Wilson SG, Zheng HF, Smith AV, Pye SR, Leo PJ, Teumer A, Hwang JY, Ohlsson C, McGuigan F, Minster RL, Hayward C, Olmos JM, Lyytikäinen LP, Lewis JR, Swart KMA, Masi L, Oldmeadow C, Holliday EG, Cheng S, van Schoor NM, Harvey NC, Kruk M, del Greco M F, Igl W, Trummer O, Grigoriou E, Luben R, Liu CT, Zhou Y, Oei L, Medina-Gomez C, Zmuda J, Tranah G, Brown SJ, Williams FM, Soranzo N, Jakobsdottir J, Siggeirsdottir K, Holliday KL, Hannemann A, Go MJ, Garcia M, Polasek O, Laaksonen M, Zhu K, Enneman AW, McEvoy M, Peel R, Sham PC, Jaworski M, Johansson Å, Hicks AA, Pludowski P, Scott R, Dhonukshe-Rutten RAM, van der Velde N, Kähönen M, Viikari JS, Sievänen H, Raitakari OT, González-Macías J, Hernández JL, Mellström D, Ljunggren O, Cho YS, Völker U, Nauck M, Homuth G, Völzke H, Haring R, Brown MA, McCloskey E, Nicholson GC, Eastell R, Eisman JA, Jones G, Reid IR, Dennison EM, Wark J, Boonen S, Vanderschueren D, Wu FCW, Aspelund T, Richards JB, Bauer D, Hofman A, Khaw KT, Dedoussis G, Obermayer-Pietsch B, Gyllensten U, Pramstaller PP, Lorenc RS, Cooper C, Kung AWC, Lips P, Alen M, Attia J, Brandi ML, de Groot LCPGM, Lehtimäki T, Riancho JA, Campbell H, Liu Y, Harris TB, Akesson K, Karlsson M, Lee JY, Wallaschofski H, Duncan EL, O'Neill TW, Gudnason V, Spector TD, Rousseau F, Orwoll E, Cummings SR, Wareham NJ, Rivadeneira F, Uitterlinden AG, Prince RL, Kiel DP, Reeve J, Kaptoge SK. Genetic determinants of heel bone properties: genome-wide association meta-analysis and replication in the GEFOS/GENOMOS consortium. Hum Mol Genet 2014; 23:3054-68. [PMID: 24430505 DOI: 10.1093/hmg/ddt675] [Show More Authors] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Quantitative ultrasound of the heel captures heel bone properties that independently predict fracture risk and, with bone mineral density (BMD) assessed by X-ray (DXA), may be convenient alternatives for evaluating osteoporosis and fracture risk. We performed a meta-analysis of genome-wide association (GWA) studies to assess the genetic determinants of heel broadband ultrasound attenuation (BUA; n = 14 260), velocity of sound (VOS; n = 15 514) and BMD (n = 4566) in 13 discovery cohorts. Independent replication involved seven cohorts with GWA data (in silico n = 11 452) and new genotyping in 15 cohorts (de novo n = 24 902). In combined random effects, meta-analysis of the discovery and replication cohorts, nine single nucleotide polymorphisms (SNPs) had genome-wide significant (P < 5 × 10(-8)) associations with heel bone properties. Alongside SNPs within or near previously identified osteoporosis susceptibility genes including ESR1 (6q25.1: rs4869739, rs3020331, rs2982552), SPTBN1 (2p16.2: rs11898505), RSPO3 (6q22.33: rs7741021), WNT16 (7q31.31: rs2908007), DKK1 (10q21.1: rs7902708) and GPATCH1 (19q13.11: rs10416265), we identified a new locus on chromosome 11q14.2 (rs597319 close to TMEM135, a gene recently linked to osteoblastogenesis and longevity) significantly associated with both BUA and VOS (P < 8.23 × 10(-14)). In meta-analyses involving 25 cohorts with up to 14 985 fracture cases, six of 10 SNPs associated with heel bone properties at P < 5 × 10(-6) also had the expected direction of association with any fracture (P < 0.05), including three SNPs with P < 0.005: 6q22.33 (rs7741021), 7q31.31 (rs2908007) and 10q21.1 (rs7902708). In conclusion, this GWA study reveals the effect of several genes common to central DXA-derived BMD and heel ultrasound/DXA measures and points to a new genetic locus with potential implications for better understanding of osteoporosis pathophysiology.
Collapse
Affiliation(s)
- Alireza Moayyeri
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Wu S, Liu Y, Zhang L, Han Y, Lin Y, Deng HW. Genome-wide approaches for identifying genetic risk factors for osteoporosis. Genome Med 2013; 5:44. [PMID: 23731620 PMCID: PMC3706967 DOI: 10.1186/gm448] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Osteoporosis, the most common type of bone disease worldwide, is clinically characterized by low bone mineral density (BMD) and increased susceptibility to fracture. Multiple genetic and environmental factors and gene-environment interactions have been implicated in its pathogenesis. Osteoporosis has strong genetic determination, with the heritability of BMD estimated to be as high as 60%. More than 80 genes or genetic variants have been implicated in risk of osteoporosis by hypothesis-free genome-wide studies. However, these genes or genetic variants can only explain a small portion of BMD variation, suggesting that many other genes or genetic variants underlying osteoporosis risk await discovery. Here, we review recent progress in genome-wide studies of osteoporosis and discuss their implications for medicine and the major challenges in the field.
Collapse
Affiliation(s)
- Shuyan Wu
- The Center for System Biomedical Research, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, No. 516 Jungong Rd, Yangpu district, Shanghai, 200093, China
| | - Yongjun Liu
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine, Tulane University, 1440 Canal St, New Orleans, LA 70112, USA
| | - Lei Zhang
- The Center for System Biomedical Research, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, No. 516 Jungong Rd, Yangpu district, Shanghai, 200093, China
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine, Tulane University, 1440 Canal St, New Orleans, LA 70112, USA
| | - Yingying Han
- The Center for System Biomedical Research, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, No. 516 Jungong Rd, Yangpu district, Shanghai, 200093, China
| | - Yong Lin
- The Center for System Biomedical Research, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, No. 516 Jungong Rd, Yangpu district, Shanghai, 200093, China
| | - Hong-Wen Deng
- The Center for System Biomedical Research, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, No. 516 Jungong Rd, Yangpu district, Shanghai, 200093, China
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine, Tulane University, 1440 Canal St, New Orleans, LA 70112, USA
| |
Collapse
|
32
|
Kwak J, Zara JN, Chiang M, Ngo R, Shen J, James AW, Le KM, Moon C, Zhang X, Gou Z, Ting K, Soo C. NELL-1 injection maintains long-bone quantity and quality in an ovariectomy-induced osteoporotic senile rat model. Tissue Eng Part A 2013; 19:426-36. [PMID: 23083222 PMCID: PMC3542871 DOI: 10.1089/ten.tea.2012.0042] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 08/24/2012] [Indexed: 02/02/2023] Open
Abstract
Over 10 million Americans have osteoporosis, and is the predominant cause of fractures in the elderly. Treatment of fractures in the setting of osteoporosis is complicated by a suboptimal bone regenerative response due to a decline in the number of osteoblasts, their function, and survival. Consequently, an osteogenic therapeutic to prevent and treat fractures in patients with osteoporosis is needed. Nel-like molecule-1 (NELL-1), a novel osteoinductive growth factor, has been shown to promote bone regeneration. In this study, we aim to demonstrate the capacity of recombinant NELL-1 to prevent ovariectomy (OVX)-induced osteoporosis in a senile rat model. Ten-month-old female Sprague-Dawley rats underwent either sham surgery or OVX. Subsequently, 50 μL of 600 μg/mL NELL-1 lyophilized onto a 0-50-μm tricalcium phosphate (TCP) carrier was injected into the femoral bone marrow cavity while phosphate-buffered saline (PBS) control was injected into the contralateral femur. Our microcomputed tomography results showed that OVX+PBS/TCP control femurs showed a continuous decrease in the bone volume (BV) and bone mineral density (BMD) from 2 to 8 weeks post-OVX. In contrast, OVX+NELL-1/TCP femurs showed resistance to OVX-induced bone resorption showing BV and BMD levels similar to that of SHAM femurs at 8 weeks post-OVX. Histology showed increased endosteal-woven bone, as well as decreased adipocytes in the bone marrow of NELL-1-treated femurs compared to control. NELL-1-treated femurs also showed increased immunostaining for bone differentiation markers osteopontin and osteocalcin. These findings were validated in vitro, in which addition of NELL-1 in OVX bone marrow stem cells resulted in increased osteogenic differentiation. Thus, NELL-1 effectively enhances in situ osteogenesis in the bone marrow, making it potentially useful in the prevention and treatment of osteoporotic fractures.
Collapse
Affiliation(s)
- Jinny Kwak
- Section of Orthodontics School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Janette N. Zara
- Dental and Craniofacial Research Institute, University of California, Los Angeles, Los Angeles, California
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California
| | - Michael Chiang
- Dental and Craniofacial Research Institute, University of California, Los Angeles, Los Angeles, California
| | - Richard Ngo
- Dental and Craniofacial Research Institute, University of California, Los Angeles, Los Angeles, California
| | - Jia Shen
- Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles, California
| | - Aaron W. James
- Section of Orthodontics School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Khoi M. Le
- Dental and Craniofacial Research Institute, University of California, Los Angeles, Los Angeles, California
| | - Crystal Moon
- Dental and Craniofacial Research Institute, University of California, Los Angeles, Los Angeles, California
| | - Xinli Zhang
- Dental and Craniofacial Research Institute, University of California, Los Angeles, Los Angeles, California
| | - Zhongru Gou
- Zhejiang-California International NanoSystems Institute, Zhejiang University, Hangzhou, China
| | - Kang Ting
- Section of Orthodontics School of Dentistry, University of California, Los Angeles, Los Angeles, California
- Dental and Craniofacial Research Institute, University of California, Los Angeles, Los Angeles, California
| | - Chia Soo
- Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
33
|
David I, Elsen JM, Concordet D. CLIP Test: a new fast, simple and powerful method to distinguish between linked or pleiotropic quantitative trait loci in linkage disequilibria analysis. Heredity (Edinb) 2012; 110:232-8. [PMID: 23250009 PMCID: PMC3668649 DOI: 10.1038/hdy.2012.70] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
An important question arises when mapping quantitative trait loci (QTLs) for genetically
correlated traits: is the correlation due to pleiotropy (a single QTL affecting more than
one trait) and/or close linkage (different QTLs that are physically close to each
other and influence the traits)? In this article, we propose the Close Linkage versus
Pleiotropism (CLIP) test, a fast, simple and powerful method to distinguish between these
two situations. The CLIP test is based on the comparison of the square of the observed
correlation between a combination of apparent effects at the marker level to the minimal
value it can take under the pleiotropic assumption. A simulation study was performed to
estimate the power and alpha risk of the CLIP test and compare it to a test that evaluated
whether the confidence intervals of the two QTLs overlapped or not (CI test). On average,
the CLIP test showed a higher power (68%) to detect close-linked QTLs than the CI
test (43%) and a same alpha risk (4%).
Collapse
Affiliation(s)
- I David
- INRA UR631 SAGA, F-31326, Castanet-Tolosan, France.
| | | | | |
Collapse
|
34
|
Abstract
During normal childhood and adolescence, the skeleton undergoes tremendous change. Utilizing the processes of modeling and remodeling, the skeleton acquires its adult configuration and ultimately achieves peak bone mass. Optimization of peak bone mass requires the proper interaction of environmental, dietary, hormonal, and genetic influences. A variety of acute and chronic conditions, as well as genetic polymorphisms, are associated with reduced bone density, which can lead to an increased risk of fracture both in childhood and later during adulthood. Bone densitometry has an established role in the evaluation of adults with bone disorders, and the development of suitable reference ranges for children now permits the application of this technology to younger individuals. We present a brief overview of the factors that determine bone density and the emerging role of bone densitometry in the assessment of bone mass in growing children and adolescents.
Collapse
Affiliation(s)
- Michael A. Levine
- Director, Center for Bone Health, Chief, Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, Professor of Pediatrics and Medicine, University of Pennsylvania Perelman School of Medicine, U.S.A.
| |
Collapse
|
35
|
Osborne DL, Weaver CM, McCabe LD, McCabe GP, Novotny R, Van Loan MD, Going S, Matkovic V, Boushey CJ, Savaiano DA. Body size and pubertal development explain ethnic differences in structural geometry at the femur in Asian, Hispanic, and white early adolescent girls living in the U.S. Bone 2012; 51:888-95. [PMID: 22944607 PMCID: PMC3491564 DOI: 10.1016/j.bone.2012.08.125] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 08/16/2012] [Accepted: 08/18/2012] [Indexed: 12/21/2022]
Abstract
UNLABELLED Variation in structural geometry is present in adulthood, but when this variation arises and what influences this variation prior to adulthood remains poorly understood. Ethnicity is commonly the focus of research of skeletal integrity and appears to explain some of the variation in quantification of bone tissue. However, why ethnicity explains variation in skeletal integrity is unclear. METHODS Here we examine predictors of bone cross sectional area (CSA) and section modulus (Z), measured using dual-energy X-ray absorptiometry (DXA) and the Advanced Hip Analysis (AHA) program at the narrow neck of the femur in adolescent (9-14 years) girls (n=479) living in the United States who were classified as Asian, Hispanic, or white if the subject was 75% of a given group based on parental reported ethnicity. Protocols for measuring height and weight follow standardized procedures. Total body lean mass (LM) and total body fat mass (FM) were quantified in kilograms using DXA. Total dietary and total dairy calcium intakes from the previous month were estimated by the use of an electronic semi-quantitative food frequency questionnaire (eFFQ). Physical activity was estimated for the previous year by a validated self-administered modifiable activity questionnaire for adolescents with energy expenditure calculated from the metabolic equivalent (MET) values from the Compendium of Physical Activities. Multiple regression models were developed to predict CSA and Z. RESULTS Age, time from menarche, total body lean mass (LM), total body fat mass (FM), height, total calcium, and total dairy calcium all shared a significant (p<0.05), positive relationship with CSA. Age, time from menarche, LM, FM, and height shared significant (p<0.05), positive relationships with Z. For both CSA and Z, LM was the most important covariate. Physical activity was not a significant predictor of geometry at the femoral neck (p≥0.339), even after removing LM as a covariate. After adjusting for covariates, ethnicity was not a significant predictor in regression models for CSA and Z. CONCLUSION Variability in bone geometry at the narrow neck of the femur is best explained by body size and pubertal maturation. After controlling for these covariates there were no differences in bone geometry between ethnic groups.
Collapse
Affiliation(s)
- D L Osborne
- Dept. of Anthropology, University of Nebraska, Lincoln, NE 68588, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Polymorphisms in the inflammatory genes CIITA, CLEC16A and IFNG influence BMD, bone loss and fracture in elderly women. PLoS One 2012; 7:e47964. [PMID: 23133532 PMCID: PMC3485004 DOI: 10.1371/journal.pone.0047964] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 09/25/2012] [Indexed: 12/26/2022] Open
Abstract
Osteoclast activity and the fine balance between bone formation and resorption is affected by inflammatory factors such as cytokines and T lymphocyte activity, mediated by major histocompatibility complex (MHC) molecules, in turn regulated by the MHC class II transactivator (MHC2TA). We investigated the effect of functional polymorphisms in the MHC2TA gene (CIITA), and two additional genes; C-type lectin domain 16A (CLEC16A), in linkage disequilibrium with CIITA and Interferon-γ (IFNG), an inducer of CIITA; on bone density, bone resorption markers, bone loss and fracture risk in 75 year-old women followed for up to 10 years (OPRA n = 1003) and in young adult women (PEAK-25 n = 999). CIITA was associated with BMD at age 75 (lumbar spine p = 0.011; femoral neck (FN) p = 0.049) and age 80 (total body p = 0.015; total hip p = 0.042; FN p = 0.028). Carriers of the CIITA rs3087456(G) allele had 1.8–3.4% higher BMD and displayed increased rate of bone loss between age 75 and 80 (FN p = 0.013; total hip p = 0.030; total body p = 3.8E−5). Despite increasing bone loss, the rs3087456(G) allele was protective against incident fracture overall (p = 0.002), osteoporotic fracture and hip fracture. Carriers of CLEC16A and IFNG variant alleles had lower BMD (p<0.05) and ultrasound parameters and a lower risk of incident fracture (CLEC16A, p = 0.011). In 25-year old women, none of the genes were associated with BMD. In conclusion, variation in inflammatory genes CIITA, CLEC-16A and INFG appear to contribute to bone phenotypes in elderly women and suggest a role for low-grade inflammation and MHC class II expression for osteoporosis pathogenesis.
Collapse
|
37
|
Hsu YH, Kiel DP. Clinical review: Genome-wide association studies of skeletal phenotypes: what we have learned and where we are headed. J Clin Endocrinol Metab 2012; 97:E1958-77. [PMID: 22965941 PMCID: PMC3674343 DOI: 10.1210/jc.2012-1890] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 07/09/2012] [Indexed: 02/07/2023]
Abstract
CONTEXT The primary goals of genome-wide association studies (GWAS) are to discover new molecular and biological pathways involved in the regulation of bone metabolism that can be leveraged for drug development. In addition, the identified genetic determinants may be used to enhance current risk factor profiles. EVIDENCE ACQUISITION There have been more than 40 published GWAS on skeletal phenotypes, predominantly focused on dual-energy x-ray absorptiometry-derived bone mineral density (BMD) of the hip and spine. EVIDENCE SYNTHESIS Sixty-six BMD loci have been replicated across all the published GWAS, confirming the highly polygenic nature of BMD variation. Only seven of the 66 previously reported genes (LRP5, SOST, ESR1, TNFRSF11B, TNFRSF11A, TNFSF11, PTH) from candidate gene association studies have been confirmed by GWAS. Among 59 novel BMD GWAS loci that have not been reported by previous candidate gene association studies, some have been shown to be involved in key biological pathways involving the skeleton, particularly Wnt signaling (AXIN1, LRP5, CTNNB1, DKK1, FOXC2, HOXC6, LRP4, MEF2C, PTHLH, RSPO3, SFRP4, TGFBR3, WLS, WNT3, WNT4, WNT5B, WNT16), bone development: ossification (CLCN7, CSF1, MEF2C, MEPE, PKDCC, PTHLH, RUNX2, SOX6, SOX9, SPP1, SP7), mesenchymal-stem-cell differentiation (FAM3C, MEF2C, RUNX2, SOX4, SOX9, SP7), osteoclast differentiation (JAG1, RUNX2), and TGF-signaling (FOXL1, SPTBN1, TGFBR3). There are still 30 BMD GWAS loci without prior molecular or biological evidence of their involvement in skeletal phenotypes. Other skeletal phenotypes that either have been or are being studied include hip geometry, bone ultrasound, quantitative computed tomography, high-resolution peripheral quantitative computed tomography, biochemical markers, and fractures such as vertebral, nonvertebral, hip, and forearm. CONCLUSIONS Although several challenges lie ahead as GWAS moves into the next generation, there are prospects of new discoveries in skeletal biology. This review integrates findings from previous GWAS and provides a roadmap for future directions building on current GWAS successes.
Collapse
Affiliation(s)
- Yi-Hsiang Hsu
- Hebrew SeniorLife Institute for Aging Research, 1200 Centre Street, Boston, Massachusetts 02131, USA
| | | |
Collapse
|
38
|
Benisch P, Schilling T, Klein-Hitpass L, Frey SP, Seefried L, Raaijmakers N, Krug M, Regensburger M, Zeck S, Schinke T, Amling M, Ebert R, Jakob F. The transcriptional profile of mesenchymal stem cell populations in primary osteoporosis is distinct and shows overexpression of osteogenic inhibitors. PLoS One 2012; 7:e45142. [PMID: 23028809 PMCID: PMC3454401 DOI: 10.1371/journal.pone.0045142] [Citation(s) in RCA: 159] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 08/13/2012] [Indexed: 12/11/2022] Open
Abstract
Primary osteoporosis is an age-related disease characterized by an imbalance in bone homeostasis. While the resorptive aspect of the disease has been studied intensely, less is known about the anabolic part of the syndrome or presumptive deficiencies in bone regeneration. Multipotent mesenchymal stem cells (MSC) are the primary source of osteogenic regeneration. In the present study we aimed to unravel whether MSC biology is directly involved in the pathophysiology of the disease and therefore performed microarray analyses of hMSC of elderly patients (79–94 years old) suffering from osteoporosis (hMSC-OP). In comparison to age-matched controls we detected profound changes in the transcriptome in hMSC-OP, e.g. enhanced mRNA expression of known osteoporosis-associated genes (LRP5, RUNX2, COL1A1) and of genes involved in osteoclastogenesis (CSF1, PTH1R), but most notably of genes coding for inhibitors of WNT and BMP signaling, such as Sclerostin and MAB21L2. These candidate genes indicate intrinsic deficiencies in self-renewal and differentiation potential in osteoporotic stem cells. We also compared both hMSC-OP and non-osteoporotic hMSC-old of elderly donors to hMSC of ∼30 years younger donors and found that the transcriptional changes acquired between the sixth and the ninth decade of life differed widely between osteoporotic and non-osteoporotic stem cells. In addition, we compared the osteoporotic transcriptome to long term-cultivated, senescent hMSC and detected some signs for pre-senescence in hMSC-OP. Our results suggest that in primary osteoporosis the transcriptomes of hMSC populations show distinct signatures and little overlap with non-osteoporotic aging, although we detected some hints for senescence-associated changes. While there are remarkable inter-individual variations as expected for polygenetic diseases, we could identify many susceptibility genes for osteoporosis known from genetic studies. We also found new candidates, e.g. MAB21L2, a novel repressor of BMP-induced transcription. Such transcriptional changes may reflect epigenetic changes, which are part of a specific osteoporosis-associated aging process.
Collapse
Affiliation(s)
- Peggy Benisch
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Tatjana Schilling
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Ludger Klein-Hitpass
- Institute of Cell Biology (Tumor Research), University Hospital Essen, Essen, Germany
| | - Sönke P. Frey
- Department of Trauma, Hand-, Plastic- and Reconstructive Surgery, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Lothar Seefried
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Nadja Raaijmakers
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Melanie Krug
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Martina Regensburger
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Sabine Zeck
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Regina Ebert
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Franz Jakob
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
- * E-mail:
| |
Collapse
|
39
|
Park JH, Song YM, Sung J, Lee K, Kim YS, Park YS. Genetic influence on bone mineral density in Korean twins and families: the healthy twin study. Osteoporos Int 2012; 23:1343-9. [PMID: 21656262 DOI: 10.1007/s00198-011-1685-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Accepted: 05/20/2011] [Indexed: 12/01/2022]
Abstract
UNLABELLED Bone mineral density (BMD), a representative marker of osteoporosis risk, is found to be highly heritable in this Korean study, which is very consistent with the findings in Western populations. This finding strongly supports that genetic factors are significant determinants of osteoporosis risk along with individual biological and behavioral factors. INTRODUCTION Although genetic factors are known to contribute significantly to variations in BMD in Western populations, such an association has not been fully evaluated in an Asian population. This study was conducted to determine the role of genetic factors on BMD in Korean population. METHODS The study participants were 2,728 men and women consisting of 497 monozygotic (MZ) twin pairs, 119 dizygotic (DZ) twin pairs, and 1,496 first-degree relatives from the Healthy Twin Study. BMD was measured using dual-energy X-ray absorptiometry. Quantitative genetic analysis based on a variance decomposition model was performed. RESULTS Age and the measured covariates accounted for 17~61% of the variation in BMD, depending on the sites of measurement. After accounting for the covariate effects, the heritability of BMD at the whole body, thoracic and lumbar spine, whole ribs, whole pelvis, whole arms, and whole legs were 0.76, 0.72, 0.73, 0.71, 0.51, and 0.75, respectively. The pair-wise correlation of BMD was the highest within MZ twin pairs, followed by DZ twin pairs, sibling pairs, and parents-child pairs. Cross-trait correlation analysis revealed a positive genetic correlation between BMDs at different sites, ranging from 0.80 (arm and leg BMD) to 0.50 (pelvis and arm BMD). CONCLUSIONS The high heritability of BMD in this Korean population similar to those found in Western populations and the significant common genetic basis between BMDs at different sites strongly supports a significant role of genetic determinants on the risk of osteoporosis.
Collapse
Affiliation(s)
- J-H Park
- Department of Family Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | | | | | | | | |
Collapse
|
40
|
Bander D, Parczewski M. Osteoporosis and vitamin D deficiency in HIV-infected patients: Genetic and classical factors compared to the HIV-associated ones – Review. HIV & AIDS REVIEW 2012. [DOI: 10.1016/j.hivar.2011.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
41
|
Shriner D. Moving toward System Genetics through Multiple Trait Analysis in Genome-Wide Association Studies. Front Genet 2012; 3:1. [PMID: 22303408 PMCID: PMC3266611 DOI: 10.3389/fgene.2012.00001] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 01/01/2012] [Indexed: 02/05/2023] Open
Abstract
Association studies are a staple of genotype–phenotype mapping studies, whether they are based on single markers, haplotypes, candidate genes, genome-wide genotypes, or whole genome sequences. Although genetic epidemiological studies typically contain data collected on multiple traits which themselves are often correlated, most analyses have been performed on single traits. Here, I review several methods that have been developed to perform multiple trait analysis. These methods range from traditional multivariate models for systems of equations to recently developed graphical approaches based on network theory. The application of network theory to genetics is termed systems genetics and has the potential to address long-standing questions in genetics about complex processes such as coordinate regulation, homeostasis, and pleiotropy.
Collapse
Affiliation(s)
- Daniel Shriner
- Center for Research on Genomics and Global Health, National Human Genome Research Institute Bethesda, MD, USA
| |
Collapse
|
42
|
Insights into the genetics of osteoporosis from recent genome-wide association studies. Expert Rev Mol Med 2011; 13:e28. [PMID: 21867596 DOI: 10.1017/s1462399411001980] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Osteoporosis, which is characterised by reduced bone mineral density (BMD) and an increased risk of fragility fractures, is the result of a complex interaction between environmental factors and genetic variants that confer susceptibility. Heritability studies have shown that BMD and other osteoporosis-related traits such as ultrasound properties of bone, skeletal geometry and bone turnover have significant inheritable components. Although previous linkage and candidate gene studies have provided few replicated loci for osteoporosis, genome-wide association approaches have produced clear and reproducible findings. To date, 20 genome-wide association studies (GWASs) for osteoporosis and related traits have been conducted, identifying dozens of genes. Further meta-analyses of GWAS data and deep resequencing of rare variants will uncover more novel susceptibility loci and ultimately provide possible therapeutic targets for fracture prevention.
Collapse
|
43
|
Hu WW, He JW, Zhang H, Wang C, Gu JM, Yue H, Ke YH, Hu YQ, Fu WZ, Li M, Liu YJ, Zhang ZL. No association between polymorphisms and haplotypes of COL1A1 and COL1A2 genes and osteoporotic fracture in postmenopausal Chinese women. Acta Pharmacol Sin 2011; 32:947-55. [PMID: 21602843 DOI: 10.1038/aps.2011.37] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
AIM To study whether genetic polymorphisms of COL1A1 and COL1A2 genes affected the onset of fracture in postmenopausal Chinese women. METHODS SNPs in COL1A1 and COL1A2 genes were identified via direct sequencing in 32 unrelated postmenopausal Chinese women. Ten SNPs were genotyped in 1252 postmenopausal Chinese women. The associations were examined using both single-SNP and haplotype tests using logistic regression. RESULTS Twenty four (4 novel) and 28 (7 novel) SNPs were identified in COL1A1 and COL1A2 gene, respectively. The distribution frequencies of 2 SNPs in COL1A1 (rs2075554 and rs2586494) and 3 SNPs in COL1A2 (rs42517, rs1801182, and rs42524) were significantly different from those documented for the European Caucasian population. No significant difference was observed between fracture and control groups with respect to allele frequency or genotype distribution in 9 selected SNPs and haplotype. No significant association was found between fragility fracture and each SNP or haplotype. The results remained the same after additional corrections for other risk factors such as weight, height, and bone mineral density. CONCLUSION Our results show no association between common genetic variations of COL1A1 and COL1A2 genes and fracture, suggesting the complex genetic background of osteoporotic fractures.
Collapse
|
44
|
Gupta M, Cheung CL, Hsu YH, Demissie S, Cupples LA, Kiel DP, Karasik D. Identification of homogeneous genetic architecture of multiple genetically correlated traits by block clustering of genome-wide associations. J Bone Miner Res 2011; 26:1261-71. [PMID: 21611967 PMCID: PMC3312758 DOI: 10.1002/jbmr.333] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Genome-wide association studies (GWAS) using high-density genotyping platforms offer an unbiased strategy to identify new candidate genes for osteoporosis. It is imperative to be able to clearly distinguish signal from noise by focusing on the best phenotype in a genetic study. We performed GWAS of multiple phenotypes associated with fractures [bone mineral density (BMD), bone quantitative ultrasound (QUS), bone geometry, and muscle mass] with approximately 433,000 single-nucleotide polymorphisms (SNPs) and created a database of resulting associations. We performed analysis of GWAS data from 23 phenotypes by a novel modification of a block clustering algorithm followed by gene-set enrichment analysis. A data matrix of standardized regression coefficients was partitioned along both axes--SNPs and phenotypes. Each partition represents a distinct cluster of SNPs that have similar effects over a particular set of phenotypes. Application of this method to our data shows several SNP-phenotype connections. We found a strong cluster of association coefficients of high magnitude for 10 traits (BMD at several skeletal sites, ultrasound measures, cross-sectional bone area, and section modulus of femoral neck and shaft). These clustered traits were highly genetically correlated. Gene-set enrichment analyses indicated the augmentation of genes that cluster with the 10 osteoporosis-related traits in pathways such as aldosterone signaling in epithelial cells, role of osteoblasts, osteoclasts, and chondrocytes in rheumatoid arthritis, and Parkinson signaling. In addition to several known candidate genes, we also identified PRKCH and SCNN1B as potential candidate genes for multiple bone traits. In conclusion, our mining of GWAS results revealed the similarity of association results between bone strength phenotypes that may be attributed to pleiotropic effects of genes. This knowledge may prove helpful in identifying novel genes and pathways that underlie several correlated phenotypes, as well as in deciphering genetic and phenotypic modularity underlying osteoporosis risk.
Collapse
Affiliation(s)
- Mayetri Gupta
- Department of Biostatistics, Boston University, Boston, MA, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Karasik D. How pleiotropic genetics of the musculoskeletal system can inform genomics and phenomics of aging. AGE (DORDRECHT, NETHERLANDS) 2011; 33:49-62. [PMID: 20596786 PMCID: PMC3063644 DOI: 10.1007/s11357-010-9159-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Accepted: 06/14/2010] [Indexed: 04/16/2023]
Abstract
Genetic study can provide insight into the biologic mechanisms underlying inter-individual differences in susceptibility to (or resistance to) organisms' aging. Recent advances in molecular genetics and genetic epidemiology provide the necessary tools to perform a study of the genetic sources of biological aging. However, to be successful, the genetic study of a complex condition requires a heritable phenotype to be developed and validated. Genome-wide association studies offer an unbiased approach to identify new candidate genes for human diseases. It is hypothesized that convergent results from multiple aging-related traits will point out the genes responsible for the general aging of the organism. This perspective focuses on the musculoskeletal aging as an example of an approach to identify a downstream common pathway that summarizes aging processes. Since the musculoskeletal traits are linked to the state of many vital functions, disability, and ultimately survival rates, we postulate that there is significance in studying musculoskeletal aging. Construction of an integrated phenotype of aging can be achieved based on shared genetics among multiple musculoskeletal biomarkers. Valid biomarkers from other systems of the organism should be similarly explored. The new composite aging score needs to be validated by determining whether it predicts all-cause mortality, incidences of major chronic diseases, and disability late in life. Comprehensive databases on biomarkers of musculoskeletal aging in multiple large cohort studies, along with information on various health outcomes, are needed to validate the proposed measure of biological aging.
Collapse
Affiliation(s)
- David Karasik
- Institute for Aging Research, Hebrew SeniorLife, Harvard Medical School, 1200 Centre Street, Boston, MA 02131, USA.
| |
Collapse
|
46
|
Karasik D, Dupuis J, Cho K, Cupples LA, Zhou Y, Kiel DP, Demissie S. Refined QTLs of osteoporosis-related traits by linkage analysis with genome-wide SNPs: Framingham SHARe. Bone 2010; 46:1114-21. [PMID: 20064633 PMCID: PMC2842472 DOI: 10.1016/j.bone.2010.01.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Revised: 12/06/2009] [Accepted: 01/02/2010] [Indexed: 11/30/2022]
Abstract
Genome-wide association studies (GWAS) using high-density array of single-nucleotide polymorphisms (SNPs) offer an unbiased strategy to identify new candidate genes for osteoporosis. We used a subset of autosomal SNPs from the Affymetrix 500K+50K SNP GeneChip marker set to examine genetic linkage with multiple highly heritable osteoporosis-related traits, including BMD of the hip and spine, heel ultrasound (attenuation and speed of sound), and geometric indices of the hip, in two generations from the Framingham Osteoporosis Study. Variance component linkage analysis was performed using normalized residuals (adjusted for age, height, BMI, and estrogen status in women). Multipoint linkage analyses produced LOD scores > or =3.0 for BMD on chromosomes (chr.) 9 and 11 and for ultrasound speed of sound on chr. 5. Hip geometric traits were linked with higher LOD scores, such as with shaft width on chr. 4 (LOD=3.9) and chr. 16 (LOD=3.8) and with shaft section modulus on chr. 22 (LOD=4.0). LOD score > or =5.0 was obtained for femoral neck width on chr. 7. In conclusion, with an SNP-based linkage approach, we identified several novel potential QTLs and confirmed previously identified chromosomal regions linked to bone mass and geometry. Subsequent focus on the spectrum of genetic polymorphisms in these refined regions may contribute to finding variants predisposing to osteoporosis.
Collapse
Affiliation(s)
- David Karasik
- Hebrew SeniorLife Institute for Aging Research and Harvard Medical School, Boston, MA 02131, USA.
| | | | | | | | | | | | | |
Collapse
|