51
|
Alonso N, Ralston SH. Unveiling the mysteries of the genetics of osteoporosis. J Endocrinol Invest 2014; 37:925-34. [PMID: 25149083 DOI: 10.1007/s40618-014-0149-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 07/28/2014] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Osteoporosis is a common disease characterised by low bone mineral density and an increased risk of fragility fractures. METHODS We conducted a literature review of relevant studies relating to the genetics of osteoporosis. RESULTS Family studies have revealed that bone density and fractures have a strong heritable component but environmental factors also play an important role. This makes identification of the causative genetic variants challenging. Linkage analysis has been successful in identifying the genes responsible for rare inherited diseases associated with abnormalities of bone mass but has been of limited value in osteoporosis. In contrast, genome-wide association studies in large cohort studies have identified 56 loci with robust evidence of association with bone density and 14 loci that predispose to fractures. Although the effect size of the implicated variants is small, many of the loci contain genes known to be involved in regulating bone cell activity through the RANK and Wnt signalling pathways, whereas others contain novel genes not previously implicated in bone metabolism. In a few instances, whole genome and exome sequencing have been successfully used to identify rare variants of large effect size that influence susceptibility to osteoporosis. CONCLUSION A future challenge will be to conduct fine mapping and functional analysis of the loci implicated in osteoporosis in order to identify the causal genetic variants and examine the mechanisms by which they influence bone cell function and bone mass. Ultimately this may lead to the identification of biomarkers for susceptibility to osteoporosis and fractures or new therapeutic targets.
Collapse
Affiliation(s)
- N Alonso
- Rheumatic Diseases Unit, Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XU, UK
| | | |
Collapse
|
52
|
Stagi S, Cavalli L, Seminara S, de Martino M, Brandi ML. The ever-expanding conundrum of primary osteoporosis: aetiopathogenesis, diagnosis, and treatment. Ital J Pediatr 2014; 40:55. [PMID: 24906390 PMCID: PMC4064514 DOI: 10.1186/1824-7288-40-55] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 05/27/2014] [Indexed: 01/07/2023] Open
Abstract
In recent years, as knowledge regarding the etiopathogenetic mechanisms of bone involvement characterizing many diseases has increased and diagnostic techniques evaluating bone health have progressively improved, the problem of low bone mass/quality in children and adolescents has attracted more and more attention, and the body evidence that there are groups of children who may be at risk of osteoporosis has grown. This interest is linked to an increased understanding that a higher peak bone mass (PBM) may be one of the most important determinants affecting the age of onset of osteoporosis in adulthood. This review provides an updated picture of bone pathophysiology and characteristics in children and adolescents with paediatric osteoporosis, taking into account the major causes of primary osteoporosis (PO) and evaluating the major aspects of bone densitometry in these patients. Finally, some options for the treatment of PO will be briefly discussed.
Collapse
Affiliation(s)
- Stefano Stagi
- Health Sciences Department, University of Florence, Anna Meyer Children's University Hospital, Florence, Italy.
| | | | | | | | | |
Collapse
|
53
|
Velázquez-Cruz R, García-Ortiz H, Castillejos-López M, Quiterio M, Valdés-Flores M, Orozco L, Villarreal-Molina T, Salmerón J. WNT3A gene polymorphisms are associated with bone mineral density variation in postmenopausal mestizo women of an urban Mexican population: findings of a pathway-based high-density single nucleotide screening. AGE (DORDRECHT, NETHERLANDS) 2014; 36:9635. [PMID: 24584697 PMCID: PMC4082595 DOI: 10.1007/s11357-014-9635-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 02/18/2014] [Indexed: 06/03/2023]
Abstract
Osteoporosis (OP) is a common skeletal disorder characterized by low bone mineral density (BMD) and is a common health problem in Mexico. To date, few genes affecting BMD variation in the Mexican population have been identified. The aim of this study was to investigate the association of single nucleotide polymorphisms (SNPs) located in genes of the Wnt pathway with BMD variation at various skeletal sites in a cohort of postmenopausal Mexican women. A total of 121 SNPs in or near 15 Wnt signaling pathway genes and 96 ancestry informative markers were genotyped in 425 postmenopausal women using the Illumina GoldenGate microarray SNP genotyping method. BMD was measured by dual-energy X-ray absorptiometry in total hip, femoral neck, Ward's triangle, and lumbar spine. Associations were tested by linear regression for quantitative traits adjusting for possible confounding factors. SNP rs752107 in WNT3A was strongly associated with decreased total hip BMD showing the highest significance under the recessive model (P = 0.00012). This SNP is predicted to disrupt a binding site for microRNA-149. In addition, a polymorphism of the Wnt antagonist DKK2 was associated with BMD in femoral neck under a recessive model (P = 0.009). Several LRP4, LRP5, and LRP6 gene variants showed site-specific associations with BMD. In conclusion, this is the first report associating Wnt pathway gene variants with BMD in the Mexican population.
Collapse
Affiliation(s)
- Rafael Velázquez-Cruz
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica, México, D.F., C.P. 14610, México,
| | | | | | | | | | | | | | | |
Collapse
|
54
|
Hu X, Shang M, Zhou J, Ye Y, Lu X, Tao C, Ying B, Wang L. Association of genetic variants in Wnt signaling pathway with tuberculosis in Chinese Han population. PLoS One 2014; 9:e93841. [PMID: 24695522 PMCID: PMC3973650 DOI: 10.1371/journal.pone.0093841] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 03/08/2014] [Indexed: 02/05/2023] Open
Abstract
Compelling studies have implicated that the Wnt signaling pathway plays an important role in the development and progression of tuberculosis, however, there is little literature addressing the role of polymorphisms in Wnt pathway on tuberculosis. We took a pathway based candidate gene approach to investigate the possible correlation between genetic variants in Wnt pathway and tuberculosis. Three single nucleotide polymorphisms (SNPs) in Wnt pathway (rs4135385 in CTNNB1 gene, rs7832767 in SFRP1 gene, and rs11079571 in AXIN2 gene) were genotyped in 422 Chinese Han tuberculosis patients and 402 frequency matched (age, gender, and ethnicity) controls using high-resolution melting analysis. The genotype and allelic frequencies of rs4135385 and rs7832767 were significantly different among patients and controls. The dominant model of rs4135385 was significantly associated with an increased risk of tuberculosis (AG/GG versus AA: OR = 1.49, 95% CI = 1.06–2.09, p = 0.019). The recessive model of rs7832767 posed a significant higher risk for tuberculosis (TT versus TC/CC, OR = 2.70, 95% CI = 1.41–5.18, p = 0.002). These SNPs were further evaluated whether they were correlated with the site of tuberculosis and the level of inflammatory markers. Rs7832767 was significantly associated with the level of CRP (p = 0.014), and the patients carrying T allele might present with elevated CRP values (OR = 1.90, 95% CI = 1.21–2.96, p = 0.005). Our study provided the first evidence that rs4135385 and rs7832767 were associated with tuberculosis risk, and genetic variants in Wnt signaling pathway might participate in genetic susceptibility to tuberculosis in Chinese Han population. Further epidemiological and functional studies in larger populations are warranted to verify our results.
Collapse
Affiliation(s)
- Xuejiao Hu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Mengqiao Shang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Juan Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Yuanxin Ye
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Xiaojun Lu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Chuanmin Tao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Binwu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
- * E-mail: (BY); (LW)
| | - Lanlan Wang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
- * E-mail: (BY); (LW)
| |
Collapse
|
55
|
Lee DO, Kim H, Ku SY, Kim SH, Kim JG. Association between polymorphisms in sclerostin, dickkopfs and secreted frizzled-related protein genes and bone mineral density in postmenopausal Korean women. Gynecol Obstet Invest 2014; 77:186-193. [PMID: 24662300 DOI: 10.1159/000358389] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 01/03/2014] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS The purpose of this study was to investigate the association between single nucleotide polymorphisms (SNPs) in sclerostin (SOST), dickkopf (DKK), secreted frizzled-related protein (sFRP) genes and bone mineral density (BMD) in postmenopausal Korean women. METHODS The SOST, Wnt inhibitory factor 1 (WIF1), sFRP1,sFRP2,sFRP3, sFRP4, sFRP5, DKK1, DKK2 and DKK3 polymorphisms were analyzed in 399 postmenopausal Korean women. Serum levels of bone turnover markers were measured, and BMDs at the lumbar spine and femoral neck were also examined. RESULTS No significant differences in adjusted BMD at the lumbar spine and femoral neck were noted according to any single and combined polymorphisms measured in SOST, DKKs and sFRPs. However, osteoporosis at the femoral neck was 2.35 times more frequently observed in the AA genotype of the sFRP4 c.958C>A polymorphism compared to the non-AA genotype (95% CI 1.09-5.08, p = 0.03). Also, the CC genotype of the sFRP3 c.970C>G polymorphism had a higher rate of osteoporosis at the femoral neck compared to the GC genotype (OR 8.47, 95% CI 1.37-52.63, p = 0.049). CONCLUSIONS Our results suggest that the sFRP3 c.970C>G and sFRP4 c.958C>A polymorphisms may be genetic factors associated with the prevalence of osteoporosis at the femoral neck in postmenopausal Korean women.
Collapse
Affiliation(s)
- Dong Ock Lee
- Department of Obstetrics and Gynecology, National Cancer Center, Koyang, Korea
| | | | | | | | | |
Collapse
|
56
|
Kuipers AL, Zhang Y, Yu S, Kammerer CM, Nestlerode CS, Chu Y, Bunker CH, Patrick AL, Wheeler VW, Miljkovic I, Zmuda JM. Relative influence of heritability, environment and genetics on serum sclerostin. Osteoporos Int 2014; 25:905-12. [PMID: 24136102 PMCID: PMC3948173 DOI: 10.1007/s00198-013-2517-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 09/16/2013] [Indexed: 01/28/2023]
Abstract
SUMMARY We determined factors associated with serum sclerostin in 446 Afro-Caribbean family members. Age, weight, sex, diabetes and kidney function were associated with sclerostin. Sclerostin was heritable, and nine SNPs in the SOST gene region were associated with sclerostin. Variation in serum sclerostin is a heritable factor that is determined by both genetic and environmental factors. INTRODUCTION Sclerostin, encoded by the SOST gene, is a Wnt inhibitor that regulates bone mineralization and is a candidate gene locus for osteoporosis. However, little is known about the genetic and non-genetic sources of inter-individual variation in serum sclerostin levels. METHODS Serum sclerostin was measured in 446 Afro-Caribbean men and women aged 18+ from seven large, multigenerational families (mean family size, 64; 3,840 relative pairs). Thirty-six common single nucleotide polymorphisms (SNP) were genotyped within a 100 kb region encompassing the gene encoding sclerostin (SOST). Genetic and non-genetic factors were tested for association with serum sclerostin. RESULTS Mean serum sclerostin was 41.3 pmol/l and was greater in men than in women (P < 0.05). Factors associated with higher serum sclerostin were increased age and body weight, male sex, diabetes and decreased glomerular filtration rate, which collectively accounted for 25.4 % of its variation. Residual genetic heritability of serum sclerostin was 0.393 (P < 0.0001). Nine SNPs reached nominal significance with sclerostin. Three of those nine SNPs represented independent association signals (rs851056, rs41455049 and rs9909172), which accounted for 7.8 % of the phenotypic variation in sclerostin, although none of these SNPs surpassed a Bonferroni correction for multiple comparisons. CONCLUSIONS Serum sclerostin is a heritable trait that is also determined by environmental factors including age, sex, adiposity, diabetes and kidney function. Three independent common SNPs within the SOST region may collectively account for a significant proportion of the variation in serum sclerostin.
Collapse
Affiliation(s)
- A L Kuipers
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
57
|
Hendrickx G, Boudin E, Fijałkowski I, Nielsen TL, Andersen M, Brixen K, Van Hul W. Variation in the Kozak sequence of WNT16 results in an increased translation and is associated with osteoporosis related parameters. Bone 2014; 59:57-65. [PMID: 24185276 DOI: 10.1016/j.bone.2013.10.022] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 10/24/2013] [Accepted: 10/25/2013] [Indexed: 11/28/2022]
Abstract
The importance of WNT16 in the regulation of bone metabolism was recently confirmed by several genome-wide association studies and by a Wnt16 (Wnt16(-/-)) knockout mouse model. The aim of this study was thus to replicate and further elucidate the effect of common genetic variation in WNT16 on osteoporosis related parameters. Hereto, we performed a WNT16 candidate gene association study in a population of healthy Caucasian men from the Odense Androgen Study (OAS). Using HapMap, five tagSNPs and one multimarker test were selected for genotyping to cover most of the common genetic variation in and around WNT16 (MAF>5%). This study confirmed previously reported associations for rs3801387 and rs2707466 with bone mineral density (BMD) at several sites. Furthermore, we additionally demonstrated that rs2908007 is strongly associated with BMD at several sites in the young, elderly and complete OAS population. The observed effect of these three associated SNPs on the respective phenotypes is comparable and we can conclude that the presence of the minor allele results in an increase in BMD. Additionally, we performed re-sequencing of WNT16 on two cohorts selected from the young OAS cohort, based on their extreme BMD values. On this basis, rs55710688 was selected for an in vitro translation experiment since it is located in the Kozak sequence of WNT16a. We observed an increased translation efficiency and thus a higher amount of WNT16a for the Kozak sequence that was significantly more prevalent in the high BMD cohort. This observation is in line with the results of the Wnt16(-/-) mice. Finally, a WNT luciferase reporter assay was performed and showed no activation of the β-catenin dependent pathway by Wnt16. We did detect a dose-dependent inhibitory effect of Wnt16 on WNT1 activation of this canonical WNT pathway. Increased translation of WNT16 can thus lead to an increased inhibitory action of WNT16 on canonical WNT signaling. This statement is in contrast with the known activating effect of canonical WNT signaling on bone formation and suggests a stimulatory effect on bone metabolism via noncanonical WNT signaling. More research is required to not only confirm this hypothesis, but also to further elucidate the role of non-canonical WNT pathways in bone metabolism and the general mechanisms of interplay between the different WNT signaling pathways.
Collapse
Affiliation(s)
- Gretl Hendrickx
- Department of Medical Genetics, University of Antwerp, Belgium.
| | - Eveline Boudin
- Department of Medical Genetics, University of Antwerp, Belgium.
| | | | | | | | - Kim Brixen
- Department of Endocrinology, Odense University Hospital, Denmark.
| | - Wim Van Hul
- Department of Medical Genetics, University of Antwerp, Belgium.
| |
Collapse
|
58
|
Chang MK, Kramer I, Keller H, Gooi JH, Collett C, Jenkins D, Ettenberg SA, Cong F, Halleux C, Kneissel M. Reversing LRP5-dependent osteoporosis and SOST deficiency-induced sclerosing bone disorders by altering WNT signaling activity. J Bone Miner Res 2014; 29:29-42. [PMID: 23901037 DOI: 10.1002/jbmr.2059] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 07/26/2013] [Accepted: 07/29/2013] [Indexed: 12/23/2022]
Abstract
The bone formation inhibitor sclerostin encoded by SOST binds in vitro to low-density lipoprotein receptor-related protein (LRP) 5/6 Wnt co-receptors, thereby inhibiting Wnt/β-catenin signaling, a central pathway of skeletal homeostasis. Lrp5/LRP5 deficiency results in osteoporosis-pseudoglioma (OPPG), whereas Sost/SOST deficiency induces lifelong bone gain in mice and humans. Here, we analyzed the bone phenotype of mice lacking Sost (Sost(-/-) ), Lrp5 (Lrp5(-/-) ), or both (Sost(-/-) ;Lrp5(-/-) ) to elucidate the mechanism of action of Sost in vivo. Sost deficiency-induced bone gain was significantly blunted in Sost(-/-) ;Lrp5(-/-) mice. Yet the Lrp5 OPPG phenotype was fully rescued in Sost(-/-) ;Lrp5(-/-) mice and most bone parameters were elevated relative to wild-type. To test whether the remaining bone increases in Sost(-/-) ;Lrp5(-/-) animals depend on Lrp6, we treated wild-type, Sost(-/-) , and Sost(-/-) ;Lrp5(-/-) mice with distinct Lrp6 function blocking antibodies. Selective blockage of Wnt1 class-mediated Lrp6 signaling reduced cancellous bone mass and density in wild-type mice. Surprisingly, it reversed the abnormal bone gain in Sost(-/-) and Sost(-/-) ;Lrp5(-/-) mice to wild-type levels irrespective of enhancement or blockage of Wnt3a class-mediated Lrp6 activity. Thus, whereas Sost deficiency-induced bone anabolism partially requires Lrp5, it fully depends on Wnt1 class-induced Lrp6 activity. These findings indicate: first, that OPPG syndrome patients suffering from LRP5 loss-of-function should benefit from principles antagonizing SOST/sclerostin action; and second, that therapeutic WNT signaling inhibitors may stop the debilitating bone overgrowth in sclerosing disorders related to SOST deficiency, such as sclerosteosis, van Buchem disease, and autosomal dominant craniodiaphyseal dysplasia, which are rare disorders without viable treatment options.
Collapse
Affiliation(s)
- Ming-Kang Chang
- Musculoskeletal Disease Area, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Tuohimaa P, Wang JH, Khan S, Kuuslahti M, Qian K, Manninen T, Auvinen P, Vihinen M, Lou YR. Gene expression profiles in human and mouse primary cells provide new insights into the differential actions of vitamin D3 metabolites. PLoS One 2013; 8:e75338. [PMID: 24116037 PMCID: PMC3792969 DOI: 10.1371/journal.pone.0075338] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 08/12/2013] [Indexed: 01/08/2023] Open
Abstract
1α,25-Dihydroxyvitamin D3 (1α,25(OH)2D3) had earlier been regarded as the only active hormone. The newly identified actions of 25-hydroxyvitamin D3 (25(OH)D3) and 24R,25-dihydroxyvitamin D3 (24R,25(OH)2D3) broadened the vitamin D3 endocrine system, however, the current data are fragmented and a systematic understanding is lacking. Here we performed the first systematic study of global gene expression to clarify their similarities and differences. Three metabolites at physiologically comparable levels were utilized to treat human and mouse fibroblasts prior to DNA microarray analyses. Human primary prostate stromal P29SN cells (hP29SN), which convert 25(OH)D3 into 1α,25(OH)2D3 by 1α-hydroxylase (encoded by the gene CYP27B1), displayed regulation of 164, 171, and 175 genes by treatment with 1α,25(OH)2D3, 25(OH)D3, and 24R,25(OH)2D3, respectively. Mouse primary Cyp27b1 knockout fibroblasts (mCyp27b1−/−), which lack 1α-hydroxylation, displayed regulation of 619, 469, and 66 genes using the same respective treatments. The number of shared genes regulated by two metabolites is much lower in hP29SN than in mCyp27b1−/−. By using DAVID Functional Annotation Bioinformatics Microarray Analysis tools and Ingenuity Pathways Analysis, we identified the agonistic regulation of calcium homeostasis and bone remodeling between 1α,25(OH)2D3 and 25(OH)D3 and unique non-classical actions of each metabolite in physiological and pathological processes, including cell cycle, keratinocyte differentiation, amyotrophic lateral sclerosis signaling, gene transcription, immunomodulation, epigenetics, cell differentiation, and membrane protein expression. In conclusion, there are three distinct vitamin D3 hormones with clearly different biological activities. This study presents a new conceptual insight into the vitamin D3 endocrine system, which may guide the strategic use of vitamin D3 in disease prevention and treatment.
Collapse
Affiliation(s)
- Pentti Tuohimaa
- Department of Anatomy, Medical School, University of Tampere, Tampere, Finland
- Department of Clinical Chemistry, Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Jing-Huan Wang
- Department of Anatomy, Medical School, University of Tampere, Tampere, Finland
- Tampere Graduate School in Biomedicine and Biotechnology, University of Tampere, Tampere, Finland
- Drug Discovery Graduate School, University of Turku, Turku, Finland
| | - Sofia Khan
- Institute of Biomedical Technology and BioMediTech, University of Tampere, Tampere, Finland
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Marianne Kuuslahti
- Department of Anatomy, Medical School, University of Tampere, Tampere, Finland
| | - Kui Qian
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Tommi Manninen
- Department of Cell Biology, Medical School, University of Tampere, Tampere, Finland
| | - Petri Auvinen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Mauno Vihinen
- Institute of Biomedical Technology and BioMediTech, University of Tampere, Tampere, Finland
- Institute of Experimental Medical Science, Lund University, Lund, Sweden
- Tampere University Hospital, Tampere, Finland
| | - Yan-Ru Lou
- Division of Biopharmaceutics and Pharmacokinetics, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- * E-mail:
| |
Collapse
|
60
|
Boudin E, Fijalkowski I, Piters E, Van Hul W. The role of extracellular modulators of canonical Wnt signaling in bone metabolism and diseases. Semin Arthritis Rheum 2013; 43:220-40. [DOI: 10.1016/j.semarthrit.2013.01.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 01/11/2013] [Accepted: 01/16/2013] [Indexed: 12/17/2022]
|
61
|
|
62
|
Low-density lipoprotein receptor-related protein 5 gene polymorphisms and genetic susceptibility to abdominal aortic aneurysm. J Vasc Surg 2013; 58:1062-8.e1. [PMID: 23490293 DOI: 10.1016/j.jvs.2012.11.092] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 11/16/2012] [Accepted: 11/22/2012] [Indexed: 11/21/2022]
Abstract
BACKGROUND Previous data showed decreased low-density lipoprotein receptor-related protein 5 (LRP5) gene expression in peripheral blood cells of abdominal aortic aneurysm (AAA) patients and an association between decreased expression of LRP5 and increased lipoprotein (a) [Lp(a)] levels in AAA. LRP5 gene is involved in bone, lipid, and glucose metabolism, and experimental studies showed that atherosclerotic lesions of ApoE:LRP5 double knockout mice were ~threefold greater than those in ApoE-knockout mice and were characterized by features of advanced atherosclerosis, with remarkable accumulation of foam cells and destruction of the internal elastic lamina. The aim of this study was to evaluate the role of polymorphisms in LRP5 gene in determining genetic susceptibility to AAA. METHODS A total of 423 AAA patients and 423 controls comparable for sex and age were genotyped for seven polymorphisms within the LRP5 (rs667126, rs3736228, rs4988300, rs3781590, rs312016, rs556442, rs627174) by TaqMan approach. RESULTS Two polymorphisms were significantly associated with AAA: rs4988300, carriers of the T allele in AAA (74.0% vs 65.3% in controls; P = .007); and rs3781590, carriers of the T allele in AAA (66.5% vs 57.4% in controls; P =.009). At the multiple logistic regression analysis, adjusted for age, sex, dyslipidemia, hypertension, smoking habit, and chronic obstructive pulmonary disease, rs4988300 and rs3781590 polymorphisms remained significant and independent determinants of AAA (OR, 1.62; 95% CI, 1.02-2.56; P = .040, and OR, 1.83; 95% CI, 1.17-2.85; P = .008, respectively). We confirmed that AAA patients had significantly higher Lp(a) levels than control subjects (180.0 mg/L vs 107.6 mg/L; P < .0001). The prevalence of patients with Lp(a) levels ≥ 300 mg/L was significantly higher in patient carriers of the rs4988300 T allele than in wild-type patients (42.6% vs 30.8%; P = .048). CONCLUSIONS Present data have identified rs4988300 and rs3781590 LPR5 polymorphisms as independent genetic markers of AAA and underlined the need to concentrate our effort in studying the role of these markers in AAA and of LRP5 gene in Lp(a) catabolism and AAA pathophysiology.
Collapse
|
63
|
WNT signaling in bone homeostasis and disease: from human mutations to treatments. Nat Med 2013; 19:179-92. [PMID: 23389618 DOI: 10.1038/nm.3074] [Citation(s) in RCA: 1520] [Impact Index Per Article: 126.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 12/18/2012] [Indexed: 12/11/2022]
Abstract
Low bone mass and strength lead to fragility fractures, for example, in elderly individuals affected by osteoporosis or children with osteogenesis imperfecta. A decade ago, rare human mutations affecting bone negatively (osteoporosis-pseudoglioma syndrome) or positively (high-bone mass phenotype, sclerosteosis and Van Buchem disease) have been identified and found to all reside in components of the canonical WNT signaling machinery. Mouse genetics confirmed the importance of canonical Wnt signaling in the regulation of bone homeostasis, with activation of the pathway leading to increased, and inhibition leading to decreased, bone mass and strength. The importance of WNT signaling for bone has also been highlighted since then in the general population in numerous genome-wide association studies. The pathway is now the target for therapeutic intervention to restore bone strength in millions of patients at risk for fracture. This paper reviews our current understanding of the mechanisms by which WNT signalng regulates bone homeostasis.
Collapse
|
64
|
Zheng G, Jinfeng X, Yuan A, Colin OW. Impact on modes of inheritance and relative risks of using extreme sampling when designing genetic association studies. Ann Hum Genet 2013; 77:80-4. [PMID: 23163532 PMCID: PMC3535545 DOI: 10.1111/j.1469-1809.2012.00733.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 08/28/2012] [Indexed: 11/29/2022]
Abstract
Using extreme phenotypes for association studies can improve statistical power . We study the impact of using samples with extremely high or low traits on the alternative model space, the genotype relative risks, and the genetic models in association studies. We prove the following results: when the risk allele causes high-trait values, the more extreme the high traits, the larger the genotype relative risks, which is not always true for using extreme low traits; we also prove that a genetic model theoretically changes with more extreme trait except for the recessive or dominant models. Practically, however, the impact of deviations from the true genetic model at a functional locus due to selective sampling is virtually negligible. The implications of our findings are discussed. Numerical values are reported for illustrations.
Collapse
Affiliation(s)
- Gang Zheng
- Office of Biostatistics Research, National Heart, Lung and Blood Institute, Bethesda, MD 20892, USA.
| | | | | | | |
Collapse
|
65
|
Boudin E, Piters E, Fijalkowski I, Stevenheydens G, Steenackers E, Kuismin O, Moilanen JS, Mortier G, Van Hul W. Mutations in sFRP1 or sFRP4 are not a common cause of craniotubular hyperostosis. Bone 2013; 52:292-5. [PMID: 23044044 DOI: 10.1016/j.bone.2012.09.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Revised: 09/26/2012] [Accepted: 09/27/2012] [Indexed: 01/10/2023]
Abstract
Sclerosing bone dysplasias are a heterogeneous group of rare diseases marked by increased BMD caused by either increased bone formation or by decreased bone resorption. In this study we have focused on craniotubular hyperostoses mainly affecting the long bones and the skull. Currently, there are three causative genes identified namely LRP5, SOST and LRP4. All three genes are involved in the canonical Wnt signalling pathway. These findings support the role of this pathway in regulating bone formation. The secreted Frizzled related proteins (sFRPs) can modulate the Wnt signalling pathway by binding to Wnt ligands or Frizzled receptors. Studies using mice showed that two members of this family, sFRP1 and sFRP4, have an important effect on bone formation. Sfrp1-/- mice have increased BMD values especially after peak BMD was reached. On the contrary, sfrp4 overexpression mice exhibit reduced BMD. Therefore, we selected sFRP1 and sFRP4, two members of the secreted Frizzled related protein (sFRP) family, as candidate genes for mutation analysis in patients with craniotubular hyperostosis. Using Sanger sequencing we screened the exons and intron/exon boundaries of sFRP1 and sFRP4 in 53 patients. In all patients mutations in LRP5, SOST and LRP4 were excluded. We identified two unknown heterozygous variants both in sFRP1. The first variant in sFRP1 is an intronic variant which, according to prediction programs, does not affect the splicing of the gene. The second variant (p.Trp131Arg/-) was identified in a young boy whose healthy mother does not carry the variant. In conclusion, our studies indicate that mutations neither in sFRP1 nor in sFRP4 are a common cause of craniotubular hyperostoses. As a consequence, further research will be necessary to identify the disease causing gene(s) in this group of patients.
Collapse
Affiliation(s)
- Eveline Boudin
- Department of Medical Genetics, University and University Hospital of Antwerp, Edegem, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
66
|
New insights into adhesion signaling in bone formation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 305:1-68. [PMID: 23890379 DOI: 10.1016/b978-0-12-407695-2.00001-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mineralized tissues that are protective scaffolds in the most primitive species have evolved and acquired more specific functions in modern animals. These are as diverse as support in locomotion, ion homeostasis, and precise hormonal regulation. Bone formation is tightly controlled by a balance between anabolism, in which osteoblasts are the main players, and catabolism mediated by the osteoclasts. The bone matrix is deposited in a cyclic fashion during homeostasis and integrates several environmental cues. These include diffusible elements that would include estrogen or growth factors and physicochemical parameters such as bone matrix composition, stiffness, and mechanical stress. Therefore, the microenvironment is of paramount importance for controlling this delicate equilibrium. Here, we provide an overview of the most recent data highlighting the role of cell-adhesion molecules during bone formation. Due to the very large scope of the topic, we focus mainly on the role of the integrin receptor family during osteogenesis. Bone phenotypes of some deficient mice as well as diseases of human bones involving cell adhesion during this process are discussed in the context of bone physiology.
Collapse
|
67
|
Zhang F, Guo X, Wu S, Han J, Liu Y, Shen H, Deng HW. Genome-wide pathway association studies of multiple correlated quantitative phenotypes using principle component analyses. PLoS One 2012; 7:e53320. [PMID: 23285279 PMCID: PMC3532454 DOI: 10.1371/journal.pone.0053320] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 11/27/2012] [Indexed: 02/07/2023] Open
Abstract
Genome-wide pathway association studies provide novel insight into the biological mechanism underlying complex diseases. Current pathway association studies primarily focus on single important disease phenotype, which is sometimes insufficient to characterize the clinical manifestations of complex diseases. We present a multi-phenotypes pathway association study(MPPAS) approach using principle component analysis(PCA). In our approach, PCA is first applied to multiple correlated quantitative phenotypes for extracting a set of orthogonal phenotypic components. The extracted phenotypic components are then used for pathway association analysis instead of original quantitative phenotypes. Four statistics were proposed for PCA-based MPPAS in this study. Simulations using the real data from the HapMap project were conducted to evaluate the power and type I error rates of PCA-based MPPAS under various scenarios considering sample sizes, additive and interactive genetic effects. A real genome-wide association study data set of bone mineral density (BMD) at hip and spine were also analyzed by PCA-based MPPAS. Simulation studies illustrated the performance of PCA-based MPPAS for identifying the causal pathways underlying complex diseases. Genome-wide MPPAS of BMD detected associations between BMD and KENNY_CTNNB1_TARGETS_UP as well as LONGEVITYPATHWAY pathways in this study. We aim to provide a applicable MPPAS approach, which may help to gain deep understanding the potential biological mechanism of association results for complex diseases.
Collapse
Affiliation(s)
- Feng Zhang
- Key Laboratory of Environment and Gene Related Diseases of Ministry Education, Faculty of Public Health, College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- * E-mail:
| | - Xiong Guo
- Key Laboratory of Environment and Gene Related Diseases of Ministry Education, Faculty of Public Health, College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Shixun Wu
- Key Laboratory of Environment and Gene Related Diseases of Ministry Education, Faculty of Public Health, College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jing Han
- Key Laboratory of Environment and Gene Related Diseases of Ministry Education, Faculty of Public Health, College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yongjun Liu
- Department of Biostatistics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, United States of America
| | - Hui Shen
- Department of Biostatistics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, United States of America
| | - Hong-Wen Deng
- Department of Biostatistics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, United States of America
| |
Collapse
|
68
|
Wang C, Zhang Z, Zhang H, He JW, Gu JM, Hu WW, Hu YQ, Li M, Liu YJ, Fu WZ, Yue H, Ke YH, Zhang ZL. Susceptibility genes for osteoporotic fracture in postmenopausal Chinese women. J Bone Miner Res 2012; 27:2582-91. [PMID: 22807154 DOI: 10.1002/jbmr.1711] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 06/22/2012] [Accepted: 07/03/2012] [Indexed: 01/01/2023]
Abstract
To identify the susceptibility genes for osteoporotic fracture in postmenopausal Chinese women, a two-stage case-control association study using joint analysis was conducted in 1046 patients with nontraumatic vertebra, hip, or distal radius fractures and 2303 healthy controls. First, 113 single-nucleotide polymorphisms (SNPs) in 16 potential osteoporosis candidate genes reported in recent genomewide association studies, meta-analyses studies, large-scale association studies, and functional studies were genotyped in a small-sample-size subgroup consisting of 541 patients with osteoporotic fractures and 554 healthy controls. Variants and haplotypes in SPTBN1, TNFRSF11B, CNR2, LRP4, and ESR1 that have been identified as being associated with osteoporotic fractures were further reanalyzed in the entire case-control group. We identified one SNP in TNFRSF11B (rs3102734), three SNPs in ESR1 (rs9397448, rs2234693, and rs1643821), two SNPs in LRP4 (rs17790156 and rs898604), and four SNPs in SPTBN1 (rs2971886, rs2941583, rs2941584, and rs12475342) were associated with all of the broadly defined osteoporotic fractures. The most significant polymorphism was rs3102734, with increased risk of osteoporotic fractures (odds ratio, 1.35; 95% confidence interval [CI], 1.17-1.55, Bonferroni p = 2.6 × 10(-4) ). Furthermore, rs3102734, rs2941584, rs12475342, rs9397448, rs2234693, and rs898604 exhibited significant allelic, genotypic, and/or haplotypic associations with vertebral fractures. SNPs rs12475342, rs9397448, and rs2234693 showed significant genotypic associations with hip fractures, whereas rs3102734, rs2073617, rs1643821, rs12475342, and rs2971886 exhibited significant genotypic and/or haplotypic associations with distal radius fractures. Accordingly, we suggest that in addition to the clinical risk factors, the variants in TNFRSF11B, SPTBN1, ESR1, and LRP4 are susceptibility genetic loci for osteoporotic fracture in postmenopausal Chinese women.
Collapse
Affiliation(s)
- Chun Wang
- Department of Osteoporosis and Bone Diseases, Metabolic Bone Disease and Genetics Research Unit, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Regard JB, Zhong Z, Williams BO, Yang Y. Wnt signaling in bone development and disease: making stronger bone with Wnts. Cold Spring Harb Perspect Biol 2012; 4:4/12/a007997. [PMID: 23209148 DOI: 10.1101/cshperspect.a007997] [Citation(s) in RCA: 145] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The skeleton as an organ is widely distributed throughout the entire vertebrate body. Wnt signaling has emerged to play major roles in almost all aspects of skeletal development and homeostasis. Because abnormal Wnt signaling causes various human skeletal diseases, Wnt signaling has become a focal point of intensive studies in skeletal development and disease. As a result, promising effective therapeutic agents for bone diseases are being developed by targeting the Wnt signaling pathway. Understanding the functional mechanisms of Wnt signaling in skeletal biology and diseases highlights how basic and clinical studies can stimulate each other to push a quick and productive advancement of the entire field. Here we review the current understanding of Wnt signaling in critical aspects of skeletal biology such as bone development, remodeling, mechanotransduction, and fracture healing. We took special efforts to place fundamentally important discoveries in the context of human skeletal diseases.
Collapse
Affiliation(s)
- Jean B Regard
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
70
|
Korvala J, Löija M, Mäkitie O, Sochett E, Jüppner H, Schnabel D, Mora S, Cole WG, Ala-Kokko L, Männikkö M. Rare variations in WNT3A and DKK1 may predispose carriers to primary osteoporosis. Eur J Med Genet 2012; 55:515-9. [DOI: 10.1016/j.ejmg.2012.06.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 06/21/2012] [Indexed: 12/30/2022]
|
71
|
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: 81] [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
|
72
|
Zheng HF, Tobias JH, Duncan E, Evans DM, Eriksson J, Paternoster L, Yerges-Armstrong LM, Lehtimäki T, Bergström U, Kähönen M, Leo PJ, Raitakari O, Laaksonen M, Nicholson GC, Viikari J, Ladouceur M, Lyytikäinen LP, Medina-Gomez C, Rivadeneira F, Prince RL, Sievanen H, Leslie WD, Mellström D, Eisman JA, Movérare-Skrtic S, Goltzman D, Hanley DA, Jones G, St. Pourcain B, Xiao Y, Timpson NJ, Smith GD, Reid IR, Ring SM, Sambrook PN, Karlsson M, Dennison EM, Kemp JP, Danoy P, Sayers A, Wilson SG, Nethander M, McCloskey E, Vandenput L, Eastell R, Liu J, Spector T, Mitchell BD, Streeten EA, Brommage R, Pettersson-Kymmer U, Brown MA, Ohlsson C, Richards JB, Lorentzon M. WNT16 influences bone mineral density, cortical bone thickness, bone strength, and osteoporotic fracture risk. PLoS Genet 2012; 8:e1002745. [PMID: 22792071 PMCID: PMC3390364 DOI: 10.1371/journal.pgen.1002745] [Citation(s) in RCA: 215] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Accepted: 04/04/2012] [Indexed: 01/29/2023] Open
Abstract
We aimed to identify genetic variants associated with cortical bone thickness (CBT) and bone mineral density (BMD) by performing two separate genome-wide association study (GWAS) meta-analyses for CBT in 3 cohorts comprising 5,878 European subjects and for BMD in 5 cohorts comprising 5,672 individuals. We then assessed selected single-nucleotide polymorphisms (SNPs) for osteoporotic fracture in 2,023 cases and 3,740 controls. Association with CBT and forearm BMD was tested for ∼2.5 million SNPs in each cohort separately, and results were meta-analyzed using fixed effect meta-analysis. We identified a missense SNP (Thr>Ile; rs2707466) located in the WNT16 gene (7q31), associated with CBT (effect size of -0.11 standard deviations [SD] per C allele, P = 6.2 × 10(-9)). This SNP, as well as another nonsynonymous SNP rs2908004 (Gly>Arg), also had genome-wide significant association with forearm BMD (-0.14 SD per C allele, P = 2.3 × 10(-12), and -0.16 SD per G allele, P = 1.2 × 10(-15), respectively). Four genome-wide significant SNPs arising from BMD meta-analysis were tested for association with forearm fracture. SNP rs7776725 in FAM3C, a gene adjacent to WNT16, was associated with a genome-wide significant increased risk of forearm fracture (OR = 1.33, P = 7.3 × 10(-9)), with genome-wide suggestive signals from the two missense variants in WNT16 (rs2908004: OR = 1.22, P = 4.9 × 10(-6) and rs2707466: OR = 1.22, P = 7.2 × 10(-6)). We next generated a homozygous mouse with targeted disruption of Wnt16. Female Wnt16(-/-) mice had 27% (P<0.001) thinner cortical bones at the femur midshaft, and bone strength measures were reduced between 43%-61% (6.5 × 10(-13)
Collapse
Affiliation(s)
- Hou-Feng Zheng
- Department of Medicine, Human Genetics, McGill University, Montreal, Canada
- Department of Epidemiology and Biostatistics, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| | - Jon H. Tobias
- Musculoskeletal Research Unit, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Emma Duncan
- Human Genetics Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, University of Queensland, Brisbane, Australia
- Endocrinology, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - David M. Evans
- Medical Research Council Centre for Causal Analyses in Translational Epidemiology, University of Bristol, Bristol, United Kingdom
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Joel Eriksson
- Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lavinia Paternoster
- Medical Research Council Centre for Causal Analyses in Translational Epidemiology, University of Bristol, Bristol, United Kingdom
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Laura M. Yerges-Armstrong
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab, University of Tampere School of Medicine and Tampere University Hospital, Tampere, Finland
| | - Ulrica Bergström
- Surgical and Perioperative Sciences, Umeå University, Umeå, Sweden
| | - Mika Kähönen
- Department of Clinical Physiology, University of Tampere School of Medicine and Tampere University Hospital, Tampere, Finland
| | - Paul J. Leo
- Human Genetics Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, University of Queensland, Brisbane, Australia
| | - Olli Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine and the Department of Clinical Physiology and Nuclear Medicine, University of Turku and Turku University Hospital, Turku, Finland
| | - Marika Laaksonen
- Department of Food and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | | | - Jorma Viikari
- Department of Medicine, University of Turku and Turku University Hospital, Turku, Finland
| | | | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab, University of Tampere School of Medicine and Tampere University Hospital, Tampere, Finland
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Richard L. Prince
- Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Perth, Australia
- School of Medicine and Pharmacology, University of Western Australia, Perth, Australia
| | | | - William D. Leslie
- Department of Internal Medicine, University of Manitoba, Winnipeg, Canada
| | - Dan Mellström
- Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - John A. Eisman
- Garvan Institute of Medical Research, University of New South Wales, Sydney, Australia
| | - Sofia Movérare-Skrtic
- Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - David Goltzman
- Department of Medicine, McGill University, Montreal, Canada
| | - David A. Hanley
- Department of Medicine, University of Calgary, Calgary, Canada
| | - Graeme Jones
- Menzies Research Institute, University of Tasmania, Hobart, Australia
| | - Beate St. Pourcain
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Yongjun Xiao
- Centre for Bone and Periodontal Research, McGill University, Montreal, Canada
| | - Nicholas J. Timpson
- Medical Research Council Centre for Causal Analyses in Translational Epidemiology, University of Bristol, Bristol, United Kingdom
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - George Davey Smith
- Medical Research Council Centre for Causal Analyses in Translational Epidemiology, University of Bristol, Bristol, United Kingdom
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Ian R. Reid
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Susan M. Ring
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Philip N. Sambrook
- Kolling Institute, Royal North Shore Hospital, University of Sydney, Sydney, Australia
| | - Magnus Karlsson
- Clinical and Molecular Osteoporosis Research Unit, Department of Orthopaedics, Skane University Hospital, Lund University, Malmö, Sweden
| | - Elaine M. Dennison
- Medical Research Council Lifecourse Epidemiology Unit, University of Southampton, Southampton, United Kingdom
| | - John P. Kemp
- Medical Research Council Centre for Causal Analyses in Translational Epidemiology, University of Bristol, Bristol, United Kingdom
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Patrick Danoy
- Human Genetics Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, University of Queensland, Brisbane, Australia
| | - Adrian Sayers
- Musculoskeletal Research Unit, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Scott G. Wilson
- Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Perth, Australia
- School of Medicine and Pharmacology, University of Western Australia, Perth, Australia
- Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Maria Nethander
- Genomics Core Facility, University of Gothenburg, Gothenburg, Sweden
| | - Eugene McCloskey
- Academic Unit of Bone Metabolism, Metabolic Bone Centre, University of Sheffield, Sheffield, United Kingdom
- NIHR Musculoskeletal Biomedical Research Unit, Sheffield Teaching Hospitals, Sheffield, United Kingdom
| | - Liesbeth Vandenput
- Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Richard Eastell
- Academic Unit of Bone Metabolism, Metabolic Bone Centre, University of Sheffield, Sheffield, United Kingdom
| | - Jeff Liu
- Lexicon Pharmaceuticals, The Woodlands, Texas, United States of America
| | - Tim Spector
- Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Braxton D. Mitchell
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Elizabeth A. Streeten
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Geriatric Research and Education Clinical Center (GRECC), Veterans Administration Medical Center, Baltimore, Maryland, United States of America
| | - Robert Brommage
- Lexicon Pharmaceuticals, The Woodlands, Texas, United States of America
| | - Ulrika Pettersson-Kymmer
- Department of Pharmacology and Neuroscience, Umeå University, Umeå, Sweden
- Department of Public Health and Clinical Medicine, Umeå Unviersity, Umeå, Sweden
| | - Matthew A. Brown
- Human Genetics Group, University of Queensland Diamantina Institute, Princess Alexandra Hospital, University of Queensland, Brisbane, Australia
| | - Claes Ohlsson
- Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - J. Brent Richards
- Department of Medicine, Human Genetics, McGill University, Montreal, Canada
- Department of Epidemiology and Biostatistics, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
- Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Mattias Lorentzon
- Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
73
|
Boudin E, Piters E, Nielsen TL, Andersen M, Roef G, Taes Y, Brixen K, Van Hul W. Single nucleotide polymorphisms in sFRP4 are associated with bone and body composition related parameters in Danish but not in Belgian men. Mol Genet Metab 2012; 106:366-74. [PMID: 22608881 DOI: 10.1016/j.ymgme.2012.04.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 04/25/2012] [Accepted: 04/25/2012] [Indexed: 11/25/2022]
Abstract
The senescence accelerated mouse P6 (SAMP6) has a low bone mass and has previously shown to be a good model for senile osteoporosis in humans. In addition to a reduced bone mass, SAMP6 mice are obese and have hyperlipidemia. Using positional cloning and expression studies, an increased expression of sfrp4 was found in these mice. SFRP4 is a modulator of the Wnt signalling pathway. This pathway has been previously shown to be involved in regulating bone mass. Additional evidence that sFRP4 has an influence on BMD was delivered by linkage and association studies mostly performed in Asian populations. Based on these data we decided to perform an association study between common variants in sFRP4, BMD, hip geometry parameters and body composition parameters in a population consisting of 1383 Danish men (783 aged 20-29 years; 600 aged 60-74 years). Afterwards we tried to replicate the significant results in a population of 994 Belgian men. In the Danish population we found 6 SNPs associated with BMD at the hip and/or femoral neck. Furthermore, all 6 SNPs were associated with several hip geometry parameters. The homozygous presence of the minor allele resulted for all SNPs (except rs4720265) in a decrease in bone density and bone strength. Finally, we observed in the Danish population age specific associations with height and fat mass. In the Belgian population we tried to replicate the results of three SNPs with BMD and body composition parameters. Unfortunately, we were not able to replicate the results found in the Danish cohort but we found one SNP (rs2598116) associated with height. In conclusion, genetic variation in sFRP4 has an influence on hip fracture risk, percentage body fat and height in a Danish male population. However, we were unable to replicate these results in an independent Belgian population.
Collapse
Affiliation(s)
- Eveline Boudin
- Department of Medical Genetics, University of Antwerp, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
74
|
Boudin E, Piters E, Fransen E, Nielsen TL, Andersen M, Roef G, Taes Y, Brixen K, Van Hul W. Association study of common variants in the sFRP1 gene region and parameters of bone strength and body composition in two independent healthy Caucasian male cohorts. Mol Genet Metab 2012; 105:508-15. [PMID: 22178351 DOI: 10.1016/j.ymgme.2011.11.189] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 11/15/2011] [Accepted: 11/15/2011] [Indexed: 12/24/2022]
Abstract
Bone mineral density (BMD) and bone strength are predictive parameters for the development of osteoporosis and related fracture later in life. Although it is well known that BMD and bone strength have a high heritability, not much of the variation is already explained. Mice models showed that sFRP1 has an influence on bone formation. Therefore this study aimed to investigate the effect of common genetic variation on BMD and bone strength in Caucasian men of different ages. Using HapMap we selected 13 tagSNPs which tag most common genetic variation in and around sFRP1 and we genotyped these SNPs in the young cohort of the Odense Androgen Study (OAS). The OAS includes a total of 1383 Danish men from two different age groups ([20-29 years]: N=783; [60-74 years]: N=600) and is well characterised. The subjects were phenotyped for BMD at several sites, and additionally for body composition and hip geometry parameters. Based on the results of the young cohort we selected three SNPs for further analysis in the complete OAS population. To conclude we tried to replicate the results of two SNPs in an independent population of 994 Belgian men. We found a strong association for rs9694405 with BMI as well in both cohorts separately as in the whole OAS population. Further we found rs4736965 associated with several hip geometry parameters in the same population. However we were not able to replicate those results in the Belgian population. At last we found in the OAS population age specific effects for rs10106678 with whole body BMD and waist to hip ratio.
Collapse
Affiliation(s)
- Eveline Boudin
- Department of Medical Genetics, University of Antwerp, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Baron R, Hesse E. Update on bone anabolics in osteoporosis treatment: rationale, current status, and perspectives. J Clin Endocrinol Metab 2012; 97:311-25. [PMID: 22238383 PMCID: PMC3275361 DOI: 10.1210/jc.2011-2332] [Citation(s) in RCA: 250] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Osteoporosis is defined as low bone mineral density associated with skeletal fractures secondary to minimal or no trauma, most often involving the spine, the hip, and the forearm. The decrease in bone mineral density is the consequence of an unbalanced bone remodeling process, with higher bone resorption than bone formation. Osteoporosis affects predominantly postmenopausal women, but also older men. This chronic disease represents a considerable medical and socioeconomic burden for modern societies. The therapeutic options for the treatment of osteoporosis have so far comprised mostly antiresorptive drugs, in particular bisphosphonates and more recently denosumab, but also calcitonin and, for women, estrogens or selective estrogen receptor modulators. These drugs have limitations, however, in particular the fact that they lead to a low turnover state where bone formation decreases with the decrease in bone-remodeling activity. In this review, we discuss the alternative class of osteoporosis drugs, i.e. bone anabolics, their biology, and the perspectives they offer for our therapeutic armamentarium. We focus on the two main osteoanabolic pathways identified as of today: PTH, the only anabolic drug currently on the market; and activation of canonical Wnt signaling through inhibition of the endogenous inhibitors sclerostin and dickkopf1. Each approach is based on a different molecular mechanism, but most recent evidence suggests that these two pathways may actually converge, at least in part. Whereas recombinant human PTH treatment is being revisited with different formulations and attempts to regulate endogenous PTH secretion via the calcium-sensing receptor, antibodies to sclerostin and dickkopf1 are currently in clinical trials and may prove to be even more efficient at increasing bone mass, possibly independent of bone turnover. Each of these anabolic approaches has its own limitations and safety issues, but the prospects of effective anabolic therapy for osteoporosis are indeed bright.
Collapse
Affiliation(s)
- Roland Baron
- Department of Medicine, Harvard Medical School, Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.
| | | |
Collapse
|
76
|
Piters E, de Freitas F, Nielsen TL, Andersen M, Brixen K, Van Hul W. Association study of polymorphisms in the SOST gene region and parameters of bone strength and body composition in both young and elderly men: data from the Odense Androgen Study. Calcif Tissue Int 2012; 90:30-9. [PMID: 22076526 DOI: 10.1007/s00223-011-9546-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 09/17/2011] [Indexed: 10/15/2022]
Abstract
By means of different genetic association studies the SOST gene, encoding sclerostin, has repeatedly been suggested to regulate bone mineral density (BMD) and osteoporosis susceptibility. This study aimed at a further understanding of the importance of two previously studied single-nucleotide polymorphisms in the SOST gene, rs10534024 (SRP3) and rs9902563 (SRP9), in the Odense Androgen Study (OAS) cohort. This cohort includes a total of 1,383 Danish men from two different age groups, 20-29 years (n = 783) and 60-74 years (n = 600), and is well characterized. Subjects were phenotyped for BMD at several sites and additionally for body composition and hip geometric parameters. In a combined analysis of the young and the elderly OAS, no associations were found for SRP3 either with BMD or with hip geometry. Instead, we found that this polymorphism had a relatively large effect on weight (-1.149 kg) and body mass index (-0.389 kg/m(2)) (P = 0.021 and 0.006 under a codominant model). For SRP9, a significant association was found for femoral neck BMD (+0.020 g/cm(2), P = 0.020) and a trend toward significance for hip geometry (buckling ratio of the narrow neck) but only when considering a recessive effect of the minor allele (C). No age-specific effects were found for either of the two SNPs. In summary, we are the first to find interesting associations between SRP3 and body composition. For SRP9, we replicated a site-specific association with femoral neck BMD. In addition, we report a novel association for this polymorphism with hip geometry.
Collapse
Affiliation(s)
- Elke Piters
- Department of Medical Genetics, University and University Hospital of Antwerp, Prins Boudewijnlaan 43B, 2650 Edegem, Belgium
| | | | | | | | | | | |
Collapse
|
77
|
Yu L, van der Valk M, Cao J, Han CYE, Juan T, Bass MB, Deshpande C, Damore MA, Stanton R, Babij P. Sclerostin expression is induced by BMPs in human Saos-2 osteosarcoma cells but not via direct effects on the sclerostin gene promoter or ECR5 element. Bone 2011; 49:1131-40. [PMID: 21890009 DOI: 10.1016/j.bone.2011.08.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 08/09/2011] [Accepted: 08/14/2011] [Indexed: 12/15/2022]
Abstract
Sclerostin is a secreted inhibitor of Wnt signaling and plays an essential role in the regulation of bone mass. The expression of sclerostin is largely restricted to osteocytes although its mode of transcriptional regulation is not well understood. We observed regulated expression of sclerostin mRNA and protein that was directly correlated with the mineralization response in cultured human Saos-2 osteosarcoma cells and rat primary calvarial cells. Sclerostin mRNA and protein levels were increased following treatment of cells with BMP2, BMP4 and BMP7. Analysis of deletion mutants from the -7.4 kb upstream region of the human sclerostin promoter did not reveal any specific regions that were responsive to BMPs, Wnt3a, PTH, TGFβ1 or Activin A in Saos-2 cells. The downstream ECR5 element did not show enhancer activity in Saos-2 cells and also was not affected when Saos-2 cells were treated with BMPs or PTH. Genome-wide microarray analysis of Saos-2 cells treated with BMP2 showed significant changes in expression of several transcription factors with putative consensus DNA binding sites in the region of the sclerostin promoter. However, whereas most factors tested showed either a range of inhibitory activity (DLX family, MSX2, HEY1, SMAD6/7) or lack of activity on the sclerostin promoter including SMAD9, only MEF2B showed a positive effect on both the promoter and ECR5 element. These results suggest that the dramatic induction of sclerostin gene expression by BMPs in Saos-2 cells occurs indirectly and is associated with late stage differentiation of osteoblasts and the mineralization process.
Collapse
MESH Headings
- Activins/pharmacology
- Adaptor Proteins, Signal Transducing
- Animals
- Bone Morphogenetic Proteins/genetics
- Bone Morphogenetic Proteins/metabolism
- Bone Morphogenetic Proteins/pharmacology
- Calcification, Physiologic/drug effects
- Calcification, Physiologic/genetics
- Cell Line, Tumor
- Enhancer Elements, Genetic/genetics
- Gene Expression Regulation, Neoplastic/drug effects
- Genetic Markers/genetics
- Humans
- Osteoblasts/drug effects
- Osteoblasts/metabolism
- Osteogenesis/drug effects
- Osteogenesis/genetics
- Osteosarcoma/genetics
- Osteosarcoma/pathology
- Parathyroid Hormone/pharmacology
- Promoter Regions, Genetic/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transforming Growth Factor beta1/pharmacology
- Wnt3A Protein/pharmacology
Collapse
Affiliation(s)
- Longchuan Yu
- Department of Metabolic Disorders, One Amgen Center Drive, Thousand Oaks, CA 91320, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Update on Wnt signaling in bone cell biology and bone disease. Gene 2011; 492:1-18. [PMID: 22079544 DOI: 10.1016/j.gene.2011.10.044] [Citation(s) in RCA: 298] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Revised: 10/13/2011] [Accepted: 10/20/2011] [Indexed: 12/17/2022]
Abstract
For more than a decade, Wnt signaling pathways have been the focus of intense research activity in bone biology laboratories because of their importance in skeletal development, bone mass maintenance, and therapeutic potential for regenerative medicine. It is evident that even subtle alterations in the intensity, amplitude, location, and duration of Wnt signaling pathways affects skeletal development, as well as bone remodeling, regeneration, and repair during a lifespan. Here we review recent advances and discrepancies in how Wnt/Lrp5 signaling regulates osteoblasts and osteocytes, introduce new players in Wnt signaling pathways that have important roles in bone development, discuss emerging areas such as the role of Wnt signaling in osteoclastogenesis, and summarize progress made in translating basic studies to clinical therapeutics and diagnostics centered around inhibiting Wnt pathway antagonists, such as sclerostin, Dkk1 and Sfrp1. Emphasis is placed on the plethora of genetic studies in mouse models and genome wide association studies that reveal the requirement for and crucial roles of Wnt pathway components during skeletal development and disease.
Collapse
|
79
|
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
|
80
|
Kim H, Choe SA, Ku SY, Kim SH, Kim JG. Association between Wnt signaling pathway gene polymorphisms and bone response to hormone therapy in postmenopausal Korean women. Menopause 2011; 18:808-813. [PMID: 21471826 DOI: 10.1097/gme.0b013e318208f9b2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The aim of this study was to explore the association between Wnt signaling pathway gene polymorphisms and response to hormone therapy (HT) as related to bone mineral density (BMD) in postmenopausal Korean women. METHODS The BMD and serum levels of osteoprotegerin, the soluble receptor activator of the nuclear factor κB ligand, and bone turnover markers were measured in 308 postmenopausal women receiving sequential estrogen + progestogen therapy. Results were analyzed according to the low-density lipoprotein receptor--related protein (LRP5) 5 c.266A > G, c.3893C > T, frizzled receptor 6 gene c.1033A > C, axin II c.148C > T, adenomatous polyposis coli c.5645T > A, and T-cell factor 1 c.766G > A polymorphisms. RESULTS The rates of 1-year changes in BMD and changes at 6 months in osteoprotegerin, soluble receptor activator of the nuclear factor κB ligand, and bone turnover markers after HT did not differ significantly between all single and haplotype genotypes of the genes studied. When a nonresponder was defined as a woman who had lost more than 3% of BMD per year after HT, women with T allele of the LRP5 c.3893C > T polymorphism showed a significantly higher risk of nonresponse at both the lumbar spine and femoral neck than did women with C allele. The risk of nonresponse at the lumbar spine was significantly higher in women with G allele of the LRP5 c.266A > G polymorphism than that in women with A allele, and the c.266G/c.3893T (GT) haplotype allele showed a similar trend. CONCLUSIONS The LRP5 c.266A > G and c.3893C > T polymorphisms may be associated with risk of nonresponse to HT in postmenopausal Korean women.
Collapse
Affiliation(s)
- Hoon Kim
- Department of Obstetrics and Gynecology, Incheon Medical Center, Seoul National University, Seoul, Korea
| | | | | | | | | |
Collapse
|
81
|
Relationship of sclerostin and secreted frizzled protein polymorphisms with bone mineral density. Menopause 2011; 18:802-7. [DOI: 10.1097/gme.0b013e3182091664] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
82
|
Duncan EL, Danoy P, Kemp JP, Leo PJ, McCloskey E, Nicholson GC, Eastell R, Prince RL, Eisman JA, Jones G, Sambrook PN, Reid IR, Dennison EM, Wark J, Richards JB, Uitterlinden AG, Spector TD, Esapa C, Cox RD, Brown SDM, Thakker RV, Addison KA, Bradbury LA, Center JR, Cooper C, Cremin C, Estrada K, Felsenberg D, Glüer CC, Hadler J, Henry MJ, Hofman A, Kotowicz MA, Makovey J, Nguyen SC, Nguyen TV, Pasco JA, Pryce K, Reid DM, Rivadeneira F, Roux C, Stefansson K, Styrkarsdottir U, Thorleifsson G, Tichawangana R, Evans DM, Brown MA. Genome-wide association study using extreme truncate selection identifies novel genes affecting bone mineral density and fracture risk. PLoS Genet 2011; 7:e1001372. [PMID: 21533022 PMCID: PMC3080863 DOI: 10.1371/journal.pgen.1001372] [Citation(s) in RCA: 194] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Accepted: 03/13/2011] [Indexed: 12/22/2022] Open
Abstract
Osteoporotic fracture is a major cause of morbidity and mortality worldwide. Low bone mineral density (BMD) is a major predisposing factor to fracture and is known to be highly heritable. Site-, gender-, and age-specific genetic effects on BMD are thought to be significant, but have largely not been considered in the design of genome-wide association studies (GWAS) of BMD to date. We report here a GWAS using a novel study design focusing on women of a specific age (postmenopausal women, age 55-85 years), with either extreme high or low hip BMD (age- and gender-adjusted BMD z-scores of +1.5 to +4.0, n = 1055, or -4.0 to -1.5, n = 900), with replication in cohorts of women drawn from the general population (n = 20,898). The study replicates 21 of 26 known BMD-associated genes. Additionally, we report suggestive association of a further six new genetic associations in or around the genes CLCN7, GALNT3, IBSP, LTBP3, RSPO3, and SOX4, with replication in two independent datasets. A novel mouse model with a loss-of-function mutation in GALNT3 is also reported, which has high bone mass, supporting the involvement of this gene in BMD determination. In addition to identifying further genes associated with BMD, this study confirms the efficiency of extreme-truncate selection designs for quantitative trait association studies.
Collapse
Affiliation(s)
- Emma L. Duncan
- University of Queensland Diamantina Institute, University of Queensland,
Princess Alexandra Hospital, Brisbane, Australia
| | - Patrick Danoy
- University of Queensland Diamantina Institute, University of Queensland,
Princess Alexandra Hospital, Brisbane, Australia
| | - John P. Kemp
- Medical Research Council Centre for Causal Analyses in Translational
Epidemiology, University of Bristol, Bristol, United Kingdom
| | - Paul J. Leo
- University of Queensland Diamantina Institute, University of Queensland,
Princess Alexandra Hospital, Brisbane, Australia
| | - Eugene McCloskey
- Academic Unit of Bone Metabolism, Metabolic Bone Centre, University of
Sheffield, Sheffield, United Kingdom
| | - Geoffrey C. Nicholson
- The University of Melbourne, Department of Clinical and Biomedical
Sciences: Barwon Health, Geelong, Australia
| | - Richard Eastell
- Academic Unit of Bone Metabolism, Metabolic Bone Centre, University of
Sheffield, Sheffield, United Kingdom
| | - Richard L. Prince
- School of Medicine and Pharmacology, University of Western Australia,
Perth, Australia
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital,
Perth, Australia
| | - John A. Eisman
- Garvan Institute of Medical Research, Sydney, Australia
- St. Vincent's Clinical School, St. Vincent's Hospital Campus,
University of New South Wales, Sydney, Australia
| | - Graeme Jones
- Menzies Research Institute, University of Tasmania, Hobart,
Australia
| | - Philip N. Sambrook
- Kolling Institute, Royal North Shore Hospital, University of Sydney,
Sydney, Australia
| | - Ian R. Reid
- Department of Medicine, University of Auckland, Auckland, New
Zealand
| | - Elaine M. Dennison
- Medical Research Council Lifecourse Epidemiology Unit, Southampton,
United Kingdom
| | - John Wark
- University of Melbourne Department of Medicine and Bone and Mineral
Service, Royal Melbourne Hospital, Melbourne, Australia
| | - J. Brent Richards
- Departments of Medicine, Human Genetics, Epidemiology and Biostatistics,
Lady Davis Institute, Jewish General Hospital, McGill University, Montreal,
Canada
- Department of Twin Research and Genetic Epidemiology, King's College
London, London, United Kingdom
| | - Andre G. Uitterlinden
- Department of Internal Medicine and Epidemiology, Erasmus Medical Center,
Rotterdam, The Netherlands
| | - Tim D. Spector
- Department of Twin Research and Genetic Epidemiology, King's College
London, London, United Kingdom
| | - Chris Esapa
- Medical Research Council Mammalian Genetics Unit, Harwell Science and
Innovation Campus, Harwell, Oxfordshire, United Kingdom
- Academic Endocrine Unit, Nuffield Department of Clinical Medicine, Oxford
Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford,
Churchill Hospital, Headington, Oxford, United Kingdom
| | - Roger D. Cox
- Medical Research Council Mammalian Genetics Unit, Harwell Science and
Innovation Campus, Harwell, Oxfordshire, United Kingdom
| | - Steve D. M. Brown
- Medical Research Council Mammalian Genetics Unit, Harwell Science and
Innovation Campus, Harwell, Oxfordshire, United Kingdom
| | - Rajesh V. Thakker
- Academic Endocrine Unit, Nuffield Department of Clinical Medicine, Oxford
Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford,
Churchill Hospital, Headington, Oxford, United Kingdom
| | - Kathryn A. Addison
- University of Queensland Diamantina Institute, University of Queensland,
Princess Alexandra Hospital, Brisbane, Australia
| | - Linda A. Bradbury
- University of Queensland Diamantina Institute, University of Queensland,
Princess Alexandra Hospital, Brisbane, Australia
| | - Jacqueline R. Center
- Garvan Institute of Medical Research, Sydney, Australia
- St. Vincent's Clinical School, St. Vincent's Hospital Campus,
University of New South Wales, Sydney, Australia
| | - Cyrus Cooper
- Medical Research Council Lifecourse Epidemiology Unit, Southampton,
United Kingdom
- National Institute for Health and Research Biomedical Research Unit,
University of Oxford, Oxford, United Kingdom
| | - Catherine Cremin
- University of Queensland Diamantina Institute, University of Queensland,
Princess Alexandra Hospital, Brisbane, Australia
| | - Karol Estrada
- Department of Internal Medicine and Epidemiology, Erasmus Medical Center,
Rotterdam, The Netherlands
| | - Dieter Felsenberg
- Centre of Muscle and Bone Research, Charité – University
Medicine Berlin, Campus Benjamin Franklin, Free and Humboldt University, Berlin,
Germany
| | - Claus-C. Glüer
- Medizinische Physik, Klinik für Diagnostische Radiologie,
Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| | - Johanna Hadler
- University of Queensland Diamantina Institute, University of Queensland,
Princess Alexandra Hospital, Brisbane, Australia
| | | | - Albert Hofman
- Department of Internal Medicine and Epidemiology, Erasmus Medical Center,
Rotterdam, The Netherlands
| | - Mark A. Kotowicz
- Department of Endocrinology and Diabetes, Barwon Health, Geelong,
Australia
| | - Joanna Makovey
- Institute of Bone Joint Research, University of Sydney, Royal North Shore
Hospital, Sydney, Australia
| | - Sing C. Nguyen
- Garvan Institute of Medical Research, Sydney, Australia
- School of Public Health and Community Medicine, University of New South
Wales, Sydney, Australia
| | - Tuan V. Nguyen
- Garvan Institute of Medical Research, Sydney, Australia
- St. Vincent's Clinical School, St. Vincent's Hospital Campus,
University of New South Wales, Sydney, Australia
- School of Public Health and Community Medicine, University of New South
Wales, Sydney, Australia
| | - Julie A. Pasco
- School of Medicine, Deakin University, Geelong, Australia
| | - Karena Pryce
- University of Queensland Diamantina Institute, University of Queensland,
Princess Alexandra Hospital, Brisbane, Australia
| | - David M. Reid
- Division of Applied Medicine, University of Aberdeen, Aberdeen, United
Kingdom
| | - Fernando Rivadeneira
- Department of Internal Medicine and Epidemiology, Erasmus Medical Center,
Rotterdam, The Netherlands
| | - Christian Roux
- Rheumatology Department, AP-HP Cochin Hospital – Paris-Descartes
University, Paris, France
| | - Kari Stefansson
- deCODE Genetics, Reykjavik, Iceland
- University of Iceland, Reykjavik, Iceland
| | | | | | - Rumbidzai Tichawangana
- The University of Melbourne, Department of Clinical and Biomedical
Sciences: Barwon Health, Geelong, Australia
| | - David M. Evans
- Medical Research Council Centre for Causal Analyses in Translational
Epidemiology, University of Bristol, Bristol, United Kingdom
| | - Matthew A. Brown
- University of Queensland Diamantina Institute, University of Queensland,
Princess Alexandra Hospital, Brisbane, Australia
- National Institute for Health and Research Biomedical Research Unit,
University of Oxford, Oxford, United Kingdom
| |
Collapse
|
83
|
Bivariate association analysis in selected samples: application to a GWAS of two bone mineral density phenotypes in males with high or low BMD. Eur J Hum Genet 2011; 19:710-6. [PMID: 21427758 DOI: 10.1038/ejhg.2011.22] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Our specific aims were to evaluate the power of bivariate analysis and to compare its performance with traditional univariate analysis in samples of unrelated subjects under varying sampling selection designs. Bivariate association analysis was based on the seemingly unrelated regression (SUR) model that allows different genetic models for different traits. We conducted extensive simulations for the case of two correlated quantitative phenotypes, with the quantitative trait locus making equal or unequal contributions to each phenotype. Our simulation results confirmed that the power of bivariate analysis is affected by the size, direction and source of the phenotypic correlations between traits. They also showed that the optimal sampling scheme depends on the size and direction of the induced genetic correlation. In addition, we demonstrated the efficacy of SUR-based bivariate test by applying it to a real Genome-Wide Association Study (GWAS) of Bone Mineral Density (BMD) values measured at the lumbar spine (LS) and at the femoral neck (FN) in a sample of unrelated males with low BMD (LS Z-scores ≤ -2) and with high BMD (LS and FN Z-scores >0.5). A substantial amount of top hits in bivariate analysis did not reach nominal significance in any of the two single-trait analyses. Altogether, our studies suggest that bivariate analysis is of practical significance for GWAS of correlated phenotypes.
Collapse
|
84
|
Novel mutations affecting LRP5 splicing in patients with osteoporosis-pseudoglioma syndrome (OPPG). Eur J Hum Genet 2011; 19:875-81. [PMID: 21407258 DOI: 10.1038/ejhg.2011.42] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Osteoporosis-pseudoglioma sydrome (OPPG) is an autosomal recessive disorder with early-onset severe osteoporosis and blindness, caused by biallelic loss-of-function mutations in the low-density lipoprotein receptor-related protein 5 (LRP5) gene. Heterozygous carriers exhibit a milder bone phenotype. Only a few splice mutations in LRP5 have been published. We present clinical and genetic data for four patients with novel LRP5 mutations, three of which affect splicing. Patients were evaluated clinically and by radiography and bone densitometry. Genetic screening of LRP5 was performed on the basis of the clinical diagnosis of OPPG. Splice aberrances were confirmed by cDNA sequencing or exon trapping. The effect of one splice mutation on LRP5 protein function was studied. A novel splice-site mutation c.1584+4A>T abolished the donor splice site of exon 7 and activated a cryptic splice site, which led to an in-frame insertion of 21 amino acids (p.E528_V529ins21). Functional studies revealed severely impaired signal transduction presumably caused by defective intracellular transport of the mutated receptor. Exon trapping was used on two samples to confirm that splice-site mutations c.4112-2A>G and c.1015+1G>T caused splicing-out of exons 20 and 5, respectively. One patient carried a homozygous deletion of exon 4 causing the loss of exons 4 and 5, as demonstrated by cDNA analysis. Our results broaden the spectrum of mutations in LRP5 and provide the first functional data on splice aberrations.
Collapse
|
85
|
Albers J, Schulze J, Beil FT, Gebauer M, Baranowsky A, Keller J, Marshall RP, Wintges K, Friedrich FW, Priemel M, Schilling AF, Rueger JM, Cornils K, Fehse B, Streichert T, Sauter G, Jakob F, Insogna KL, Pober B, Knobeloch KP, Francke U, Amling M, Schinke T. Control of bone formation by the serpentine receptor Frizzled-9. ACTA ACUST UNITED AC 2011; 192:1057-72. [PMID: 21402791 PMCID: PMC3063134 DOI: 10.1083/jcb.201008012] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Although Wnt signaling in osteoblasts is of critical importance for the regulation of bone remodeling, it is not yet known which specific Wnt receptors of the Frizzled family are functionally relevant in this process. In this paper, we show that Fzd9 is induced upon osteoblast differentiation and that Fzd9(-/-) mice display low bone mass caused by impaired bone formation. Our analysis of Fzd9(-/-) primary osteoblasts demonstrated defects in matrix mineralization in spite of normal expression of established differentiation markers. In contrast, we observed a reduced expression of chemokines and interferon-regulated genes in Fzd9(-/-) osteoblasts. We also identified the ubiquitin-like modifier Isg15 as one potential downstream mediator of Fzd9 in these cells. Importantly, our molecular analysis further revealed that canonical Wnt signaling is not impaired in the absence of Fzd9, thus explaining the absence of a bone resorption phenotype. Collectively, our results reveal a previously unknown function of Fzd9 in osteoblasts, a finding that may have therapeutic implications for bone loss disorders.
Collapse
Affiliation(s)
- Joachim Albers
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Rojo Venegas K, Aguilera Gómez M, Eisman JA, García Sánchez A, Faus Dader MJ, Calleja Hernández MA. Pharmacogenetics of osteoporosis-related bone fractures: moving towards the harmonization and validation of polymorphism diagnostic tools. Pharmacogenomics 2011; 11:1287-303. [PMID: 20860468 DOI: 10.2217/pgs.10.116] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Osteoporosis is one of the most common skeletal chronic conditions in developed countries, hip fracture being one of its major healthcare outcomes. There is considerable variation in the implementation of current pharmacological treatment and prevention, despite consistent recommendations and guidelines. Many studies have reported conflicting findings of genetic associations with bone density and turnover that might predict fracture risk. Moreover, it is not clear whether genetic differences exist in relation to the morbidity and efficiency of the pharmacotherapy treatments. Clinical response, including beneficial and adverse events associated with osteoporosis treatments, is highly variable among individuals. In this context, the present article intends to summarize putative candidate genes and genome-wide association studies that have been related with BMD and fracture risk, and to draw the attention to the need for pharmacogenetic methodology that could be applicable in clinical translational research after an adequate validation process. This article mainly compiles analysis of important polymorphisms in osteoporosis documented previously, and it describes the simple molecular biology tools for routine genotype acquisition. Validation of methods for the easy, fast and accessible identification of SNPs is necessary for evolving pharmacogenetic diagnostic tools in order to contribute to the discovery of clinically relevant genetic variation with an impact on osteoporosis and its personalized treatment.
Collapse
Affiliation(s)
- Karen Rojo Venegas
- Pharmacogenetics Unit, Pharmacy Service, University Hospital Virgen de las Nieves, Avenida de las Fuerzas Armadas 2, CP:18014, Granada, Spain.
| | | | | | | | | | | |
Collapse
|
87
|
Riancho JA, Olmos JM, Pineda B, García-Ibarbia C, Pérez-Núñez MI, Nan DN, Velasco J, Cano A, García-Pérez MA, Zarrabeitia MT, González-Macías J. Wnt receptors, bone mass, and fractures: gene-wide association analysis of LRP5 and LRP6 polymorphisms with replication. Eur J Endocrinol 2011; 164:123-31. [PMID: 20926594 DOI: 10.1530/eje-10-0582] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Genes explaining the susceptibility to osteoporosis have not been fully elucidated. Our objective was to explore the association of polymorphisms capturing common variations of the lipoprotein receptor-related protein (LRP) 5 and 6 genes, encoding two Wnt receptors, with femoral neck bone mineral density (BMD) and osteoporotic fractures of the spine and the hip. DESIGN Cross-sectional, case-control, and replication genetic association study. METHODS Thirty-nine tagging and functional single nucleotide polymorphisms (SNPs) were analyzed in a group of 1043 postmenopausal women and 394 women with hip fractures. The results were replicated in a different group of 342 women. RESULTS Three SNPs of the LRP6 gene were associated with BMD (nominal uncorrected P values <0.05) in the discovery cohort. One showed a significant association after multiple test correction; two of them were also associated in the replication cohort, with a combined standardized mean difference of 0.51 (P=0.009) and 0.47 (P<0.003) across rs11054704 and rs2302685 genotypes. In the discovery cohort, several LRP5 SNPs were associated with vertebral fractures (odds ratio (OR) 0.67; P=0.01), with hip fractures (unadjusted ORs between 0.59 and 1.21; P=0.005-0.033, but not significant after multiple test adjustment or age adjustment), and with height and the projected femoral neck area, but not with BMD. Transcripts of LRP5 and LRP6 were similarly abundant in bone samples. CONCLUSIONS In this study, we found common polymorphisms of LRP5 associated with osteoporotic fractures, and polymorphisms of the LRP6 gene associated with BMD, thus suggesting them as likely candidates to contribute to the explaination of the hereditary influence on osteoporosis.
Collapse
Affiliation(s)
- José A Riancho
- Department of Internal Medicine, Hospital U.M. Valdecilla-IFIMAV, University of Cantabria, RETICEF, Avda Valdecilla s/n, 39008 Santander, Spain.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Li WF, Hou SX, Yu B, Jin D, Férec C, Chen JM. Genetics of osteoporosis: perspectives for personalized medicine. Per Med 2010; 7:655-668. [PMID: 29788568 DOI: 10.2217/pme.10.55] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Osteoporosis is the most common metabolic bone disorder worldwide. At least 15 genes (e.g., ESR1, LRP5, SOST, OPG, RANK and RANKL) have been confirmed as osteoporosis susceptibility genes, and another 30 have been highlighted as promising susceptibility genes. Notably, these genes are clustered in three biological pathways: the estrogen endocrine pathway, the Wnt/β-catenin signaling pathway and the RANK/RANKL/osteoprotegerin (OPG) pathway. In this article, using data pertaining to these three biological pathways as examples, we illustrate possible principles of personalized therapy for osteoporosis. In particular, we propose to use inhibitors (e.g., denosumab) of the RANK/RANKL/OPG signaling pathway to circumvent resistance to estrogen-replacement therapy: a novel idea resulting from the consideration of a mechanistic link between the estrogen endocrine pathway and the RANK/RANKL/OPG signaling pathway. In addition, we call for more attention to be focused on rare variants of major effects in future studies.
Collapse
Affiliation(s)
- Wen-Feng Li
- Department of Orthopaedics, The First Affiliated Hospital, General Hospital of the People’s Liberation Army, Beijing, China
| | - Shu-Xun Hou
- Department of Orthopaedics, The First Affiliated Hospital, General Hospital of the People’s Liberation Army, Beijing, China
| | - Bin Yu
- Department of Orthopaedic Trauma, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dan Jin
- Department of Orthopaedic Trauma, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Claude Férec
- Institut National de la Santé et de la Recherche Médicale (INSERM), U613, Brest, France; INSERM, U613 and EFS – Bretagne, 46 rue Félix Le Dantec, 29218 Brest, France
- Etablissement Français du Sang (EFS) – Bretagne, Brest, France
- Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale (UBO), Brest, France
| | | |
Collapse
|
89
|
Lee DY, Kim H, Ku SY, Kim SH, Choi YM, Kim JG. Association between polymorphisms in Wnt signaling pathway genes and bone mineral density in postmenopausal Korean women. Menopause 2010; 17:1064-1070. [PMID: 20613673 DOI: 10.1097/gme.0b013e3181da4da3] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The purpose of this study was to investigate the association between single nucleotide polymorphisms in Wnt signal pathway genes and circulating osteoprotegerin (OPG), soluble receptor activator of the nuclear factor-κB ligand (sRANKL) levels, bone turnover markers, and bone mineral density (BMD) in postmenopausal Korean women. METHODS Wnt9a c256G>A; low-density lipoprotein receptor-related protein (LRP) 5 c266A>G, c2245C>G, c3893C>T, and c4099G>A; secreted frizzled-related protein (sFRP) 4 c1019G>A; axin II c148C>T and c1615G>A; glycogen synthase kinase binding protein (GBP) c455C>A; β-catenin c94G>T and c101G>T; T-cell factor 1 c663G>T, c734C>T, and c766G>A; and adenomatous polyposis coli c5465T>A polymorphisms were analyzed in 392 postmenopausal Korean women. Serum levels of OPG, sRANKL, and bone turnover markers were measured, and BMDs at the lumbar spine and femoral neck were examined. RESULTS Wnt9a c256G>A, LRP5 c2245C>G and c4099G>A, axin II c1615G>A, GBP c455C>A, β-catenin c94G>T and c101G>T, and T-cell factor 1 c663G>T and c734C>T single nucleotide polymorphisms were not observed. Among the genes showing polymorphisms, only the sFRP4 c1019G>A polymorphism was associated with BMD. The AA genotype in the sFRP4 c1019G>A polymorphism showed significantly lower lumbar spine BMD and a higher serum bone alkaline phosphatase level than did the GG genotype and showed a 6.39 times higher risk for osteoporosis at the lumbar spine compared with the GG genotype. No significant differences in bone turnover markers, OPG, and sRANKL were detected among the other single genotypes or the LRP haplotype genotype. CONCLUSIONS Our results suggest that the sFRP4 c1019G>A polymorphism may be one of the genetic factors affecting lumbar spine BMD in postmenopausal Korean women.
Collapse
Affiliation(s)
- Dong-Yun Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
90
|
Esteve P, Bovolenta P. The advantages and disadvantages of sfrp1 and sfrp2 expression in pathological events. TOHOKU J EXP MED 2010; 221:11-7. [PMID: 20448436 DOI: 10.1620/tjem.221.11] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Secreted Frizzled Related Proteins (Sfrps) are a family of secreted proteins that can bind both to Wnt ligands and Frizzled receptors, thereby modulating the Wnt signalling cascades. Recent studies have shown that Sfrps can also interact with Wnt unrelated molecules such as RANKL, a member of the tumor necrosis factor family, Tolloid metalloproteinases or integrin-fibronectin complexes. Alterations in the levels of Sfrp expression have been recently associated with different pathological conditions, including tumor formation and bone and myocardial disorders. Here, we summarise the evidence that relates Sfrps with these diseases and discuss how the proposed multiple Sfrp interactions with Wnt related and unrelated pathways may explain their implication in such diverse pathologies.
Collapse
Affiliation(s)
- Pilar Esteve
- Departamento de Neurobiología Molecular, Celular y del Desarrollo, Instituto Cajal (CSIC), Spain.
| | | |
Collapse
|
91
|
Abstract
Osteoporosis is an important and complex disorder that is highly prevalent worldwide. This disease poses a major challenge to modern medicine and its treatment is associated with high costs. Numerous studies have endeavored to decipher the pathogenesis of this disease. The clinical assessment of patients often incorporates information about a family history of osteoporotic fractures. Indeed, the observation of an increased risk of fracture in an individual with a positive parental history of hip fracture provides strong evidence for the heritability of osteoporosis. The onset and progression of osteoporosis are generally controlled by multiple genetic and environmental factors, as well as interactions between them, with rare cases determined by a single gene. In an attempt to identify the genetic markers of complex diseases such as osteoporosis, there has been a move away from traditional linkage mapping studies and candidate gene association studies to higher-density genome-wide association studies. The advent of high-throughput technology enables genotyping of millions of DNA markers in the human genome, and consequently the identification and characterization of causal variants and loci that underlie osteoporosis. This Review presents an overview of the major findings since 2007 and clinical applications of these genome-wide linkage and association studies.
Collapse
|
92
|
Xu XH, Dong SS, Guo Y, Yang TL, Lei SF, Papasian CJ, Zhao M, Deng HW. Molecular genetic studies of gene identification for osteoporosis: the 2009 update. Endocr Rev 2010; 31:447-505. [PMID: 20357209 PMCID: PMC3365849 DOI: 10.1210/er.2009-0032] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Accepted: 02/02/2010] [Indexed: 12/12/2022]
Abstract
Osteoporosis is a complex human disease that results in increased susceptibility to fragility fractures. It can be phenotypically characterized using several traits, including bone mineral density, bone size, bone strength, and bone turnover markers. The identification of gene variants that contribute to osteoporosis phenotypes, or responses to therapy, can eventually help individualize the prognosis, treatment, and prevention of fractures and their adverse outcomes. Our previously published reviews have comprehensively summarized the progress of molecular genetic studies of gene identification for osteoporosis and have covered the data available to the end of September 2007. This review represents our continuing efforts to summarize the important and representative findings published between October 2007 and November 2009. The topics covered include genetic association and linkage studies in humans, transgenic and knockout mouse models, as well as gene-expression microarray and proteomics studies. Major results are tabulated for comparison and ease of reference. Comments are made on the notable findings and representative studies for their potential influence and implications on our present understanding of the genetics of osteoporosis.
Collapse
Affiliation(s)
- Xiang-Hong Xu
- Institute of Molecular Genetics, Xi'an Jiaotong University, Shaanxi, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
93
|
Lauretani F, Cepollaro C, Bandinelli S, Cherubini A, Gozzini A, Masi L, Falchetti A, Del Monte F, Carbonell-Sala S, Marini F, Tanini A, Corsi AM, Ceda GP, Brandi ML, Ferrucci L. LRP5 gene polymorphism and cortical bone. Aging Clin Exp Res 2010; 22:281-8. [PMID: 21116122 PMCID: PMC5139676 DOI: 10.1007/bf03324935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND AIMS There is evidence that distinct genetic polymorphisms of LRP5 are associated with low Bone Mineral Density (BMD) and the risk of fracture. However, relationships between LRP5 polymorphisms and micro- and macro architectural bone characteristics assessed by pQCT have not been studied. The aim of the present study was to investigate the association of Ala1330Val and Val667Met polymorphisms in LRP5 gene with volumetric BMD (vBMD) and macro-architectural bone parameters in a population-based sample of men and women. METHODS We studied 959 participants of the InCHIANTI study (451 men and 508 women, age range: 21-94 yrs). Trabecular vBMD (vBMDt, mg/cm3), cortical vBMD (vBMDc, mg/cm3), cortical bone area (CBA, mm2) and cortical thickness (Ct.Th, mm) at the level of the tibia were assessed by peripheral quantitative computed tomography (pQCT). Ala1330Val and Val667Met genotypes were determined on genomic DNA by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). RESULTS In age-adjusted analyses both LRP 1330-valine and LRP 667-metionin variants were associated with lower vBMDt in men (p<0.05), and lower vBMDt (p<0.05), Ct.Th (p<0.05) and CBA (p<0.05) in women. After adjusting for multiple confounders, only the association of LRP5 1330-valine and 667-metionin with CBA remained statistically significant (p=0.04 and p=0.01, respectively) in women. CONCLUSION These findings suggest that both Ala1330Val and Val667Met LRP5 polymorphisms may affect the determination of geometric bone parameters in women.
Collapse
|
94
|
Agueda L, Urreizti R, Bustamante M, Jurado S, Garcia-Giralt N, Díez-Pérez A, Nogués X, Mellibovsky L, Grinberg D, Balcells S. Analysis of three functional polymorphisms in relation to osteoporosis phenotypes: replication in a Spanish cohort. Calcif Tissue Int 2010; 87:14-24. [PMID: 20390408 DOI: 10.1007/s00223-010-9361-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2009] [Accepted: 03/24/2010] [Indexed: 01/11/2023]
Abstract
Osteoporosis is a complex disease involving many putative genetic factors. Association analysis of functional SNPs in candidate genes is an important tool for their identification. However, this approach is affected by limited power, population stratification, and other drawbacks that lead to discordant results. Replication in independent cohorts is essential. We performed association analyses of three functional polymorphisms previously associated with bone phenotypes--namely, Ala222Val in MTHFR, Ile1062Val in LRP6, and -13910C>T in LCT--in a cohort of 944 postmenopausal Spanish women, all of them with lumbar spine (LS) bone mineral density (BMD) data and most with femoral neck (FN) BMD and fracture data. We found significant differences between genotypes only for the MTHFR polymorphism and vertebral factures, with an OR of 2.27 (95% CI 1.17-4.38) for the TT vs. CC/CT genotypes, P = 0.018. We present genotype and allele frequency data for LCT -13910C>T for a Spanish population, where the T allele (conferring lactase persistence) has a frequency of 38.6%. Genotype frequencies were consistent with observed clines in Europe and with the prevalence of lactase nonpersistence. The LCT -13910C>T polymorphism was significantly associated with height and weight, such that T allele carriers were 0.88 cm taller (95% CI 0.08-1.59 cm, P = 0.032, adjusted by age) than CC individuals and TT homozygotes were 1.91 kg heavier than CC/CT individuals (95% CI 0.11-3.71 kg, P = 0.038, adjusted by age). In conclusion, no significant association was observed between the studied polymorphisms and LS BMD or FN BMD in postmenopausal Spanish women, and only MTHFR Ala222Val was associated with vertebral fractures.
Collapse
Affiliation(s)
- Lídia Agueda
- Department of Genetics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Duncan EL, Brown MA. Clinical review 2: Genetic determinants of bone density and fracture risk--state of the art and future directions. J Clin Endocrinol Metab 2010; 95:2576-87. [PMID: 20375209 DOI: 10.1210/jc.2009-2406] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
CONTEXT Osteoporosis is a common, highly heritable condition that causes substantial morbidity and mortality, the etiopathogenesis of which is poorly understood. Genetic studies are making increasingly rapid progress in identifying the genes involved. EVIDENCE ACQUISITION AND SYNTHESIS In this review, we will summarize the current understanding of the genetics of osteoporosis based on publications from PubMed from the year 1987 onward. CONCLUSIONS Most genes involved in osteoporosis identified to date encode components of known pathways involved in bone synthesis or resorption, but as the field progresses, new pathways are being identified. Only a small proportion of the total genetic variation involved in osteoporosis has been identified, and new approaches will be required to identify most of the remaining genes.
Collapse
Affiliation(s)
- Emma L Duncan
- University of Queensland Diamantina Institute for Cancer, Immunology and Metabolic Medicine, Princess Alexandra Hospital, Ipswich Road, Woolloongabba, Queensland 4102, Australia.
| | | |
Collapse
|
96
|
Duncan EL, Brown MA. Mapping genes for osteoporosis--old dogs and new tricks. Bone 2010; 46:1219-25. [PMID: 20060943 DOI: 10.1016/j.bone.2009.12.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Revised: 12/27/2009] [Accepted: 12/30/2009] [Indexed: 12/15/2022]
Abstract
In stark contrast to its horticultural origins, modern genetics is an extremely technology-driven field. Almost all the major advances in the field over the past 20 years have followed technological developments that have permitted change in study designs. The development of PCR in the 1980s led to RFLP mapping of monogenic diseases. The development of fluorescent-tagged genotyping methods led to linkage mapping approaches for common diseases that dominated the 1990s. The development of microarray SNP genotyping has led to the genome-wide association study era of the new millennium. And now the development of next-generation sequencing technologies is about to open up a new era of gene-mapping, enabling many potential new study designs. This review aims to present the strengths and weaknesses of the current approaches, and present some new ideas about gene-mapping approaches that are likely to advance our knowledge of the genes involved in heritable bone traits such as bone mineral density (BMD) and fracture.
Collapse
Affiliation(s)
- Emma L Duncan
- Diamantina Institute of Cancer, Immunology and Metabolic Medicine, Princess Alexandra Hospital, Ipswich Road, Woolloongabba, QLD 4102, Australia
| | | |
Collapse
|
97
|
Piters E, Balemans W, Nielsen TL, Andersen M, Boudin E, Brixen K, Van Hul W. Common genetic variation in the DKK1 gene is associated with hip axis length but not with bone mineral density and bone turnover markers in young adult men: results from the Odense Androgen Study. Calcif Tissue Int 2010; 86:271-81. [PMID: 20101398 DOI: 10.1007/s00223-010-9334-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2009] [Accepted: 01/02/2010] [Indexed: 12/17/2022]
Abstract
LRP5 was recently confirmed as an important susceptibility gene for osteoporosis. Our objective was to evaluate the effect of DKK1 polymorphisms on bone mineral density (BMD), hip geometry, and bone turnover. DKK1 is a secreted protein that binds to LRP5/6 receptors and inhibits canonical Wnt signaling. Using HapMap, we selected three SNPs covering the genetic variation in a 13.53-kb region comprising DKK1. The Odense Androgen Study is a population-based study comprising 783 Caucasian men aged 20-29 years. BMD and hip structural parameters were available for study. Bone turnover markers were used as a secondary end point. All analyses were repeated after adjusting for covariables and in subgroups according to physical activity. We found no significant association between DKK1 and BMD or markers of bone turnover; however, a significant association (P = 0.012) was found for rs1569198 with hip axis length (HAL), independent of BMD and height. Moreover, the association seemed to be driven by the non-sedentary subgroup (P = 0.004). Haplotype analysis further confirmed the association of rs1569198 with HAL. Furthermore, we obtained indications for interaction between DKK1 and LRP5 genotypes for different hip geometry parameters. As almost all variance within the DKK1 gene was covered, we conclude that common variation in this gene does not markedly influence BMD or bone turnover markers in young men. In this population, however, a common SNP in DKK1 does have a significant effect on HAL, implying a possible effect on hip fracture risk in the general population. This finding could be of interest but needs replication in independent populations.
Collapse
Affiliation(s)
- Elke Piters
- Department of Medical Genetics, University of Antwerp and University Hospital, Antwerp, Belgium.
| | | | | | | | | | | | | |
Collapse
|
98
|
Reppe S, Refvem H, Gautvik VT, Olstad OK, Høvring PI, Reinholt FP, Holden M, Frigessi A, Jemtland R, Gautvik KM. Eight genes are highly associated with BMD variation in postmenopausal Caucasian women. Bone 2010; 46:604-12. [PMID: 19922823 DOI: 10.1016/j.bone.2009.11.007] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Revised: 10/22/2009] [Accepted: 11/09/2009] [Indexed: 02/09/2023]
Abstract
Low bone mineral density (BMD) is an important risk factor for skeletal fractures which occur in about 40% of women >/=50 years in the western world. We describe the transcriptional changes in 84 trans-iliacal bone biopsies associated with BMD variations in postmenopausal females (50 to 86 years), aiming to identify genetic determinants of bone structure. The women were healthy or having a primary osteopenic or osteoporotic status with or without low energy fractures. The total cohort of 91 unrelated women representing a wide range of BMDs, were consecutively registered and submitted to global gene Affymetrix microarray expression analysis or histomorphometry. Among almost 23,000 expressed transcripts, a set represented by ACSL3 (acyl-CoA synthetase long-chain family member 3), NIPSNAP3B (nipsnap homolog 3B), DLEU2 (Deleted in lymphocytic leukemia, 2), C1ORF61 (Chromosome 1 open reading frame 61), DKK1 (Dickkopf homolog 1), SOST (Sclerostin), ABCA8, (ATP-binding cassette, sub-family A, member 8), and uncharacterized (AFFX-M27830-M-at), was significantly correlated to total hip BMD (5% false discovery rate) explaining 62% of the BMD variation expressed as T-score, 53% when adjusting for the influence of age (Z-score) and 44% when further adjusting for body mass index (BMI). Only SOST was previously associated to BMD, and the majority of the genes have previously not been associated with a bone phenotype. In molecular network analyses, SOST shows a strong, positive correlation with DKK1, both being members of the Wnt signaling pathway. The results provide novel insight in the underlying biology of bone metabolism and osteoporosis which is the ultimate consequence of low BMD.
Collapse
Affiliation(s)
- Sjur Reppe
- Institute of Basic Medical Sciences, University of Oslo, Norway.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Affiliation(s)
- Natalie A Sims
- St. Vincent's Institute and Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, Victoria, Australia
| |
Collapse
|
100
|
Ohnaka K, Yamamoto K, Nakamura K, Adachi M, Kawate H, Kono S, Takayanagi R. Association of single nucleotide polymorphisms in secreted frizzled-related protein 1 gene with bone mineral density in Japanese women. Geriatr Gerontol Int 2010; 9:304-9. [PMID: 19702942 DOI: 10.1111/j.1447-0594.2009.00540.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
AIM Recent studies have demonstrated that the Wnt signaling pathway plays an important role in bone metabolism. The purpose of this study was to examine whether the gene of secreted frizzled-related protein 1 (SFRP1), a Wnt antagonist, is involved in the etiology of osteoporosis using association study. METHODS Seven single nucleotide polymorphisms (SNP) in the SFRP1 gene were genotyped and analyzed for association with bone mineral density (BMD) in 931 Japanese women (63.5 +/- 6.7 years old, mean +/- standard deviation). RESULTS One SNP (rs16890444) located in intron and another (rs3242) located in the 3'-untranslated region of the sFRP1 gene were significantly associated with the lumbar spine BMD value, and BMD values for both the femoral neck and the total hip, respectively. Women with the T/T genotype of the former SNP had a lower BMD value of the lumbar spine (L2-L4) compared with those with C/C or C/T (BMD value adjusted for age, duration after menopause, and body mass index: 0.781 vs 0.830, P = 0.037), while women with the T/T genotype of the latter SNP had higher BMD values of femoral neck and total hip compared with those with C/C or C/T (adjusted BMD value: femoral neck, 0.721 vs 0.633, P = 0.025; total hip, 0.834 vs 0.737, P = 0.027). CONCLUSION These results suggest that the SFRP1 may be a candidate gene for a BMD determinant, but further studies need to consolidate the present findings.
Collapse
Affiliation(s)
- Keizo Ohnaka
- Department of Geriatric Medicine, Kyorin University School of Medicine, Mitaka, Japan
| | | | | | | | | | | | | |
Collapse
|