1
|
Punt LD, Kooijman S, Mutsters NJM, Yue K, van der Kaay DCM, van Tellingen V, Bakker-van Waarde WM, Boot AM, van den Akker ELT, van Boekholt AA, de Groote K, Kruijsen AR, van Nieuwaal-van Maren NHG, Woltering MC, Heijligers M, van der Heyden JC, Bannink EMN, Rinne T, Hannema SE, de Waal WJ, Delemarre LC, Rensen PCN, de Bruin C, van Duyvenvoorde HA, Visser JA, Delhanty PJD, Losekoot M, Wit JM, Joustra SD. Loss-of-Function GHSR Variants Are Associated With Short Stature and Low IGF-I. J Clin Endocrinol Metab 2025; 110:e1303-e1314. [PMID: 39785833 PMCID: PMC12012706 DOI: 10.1210/clinem/dgaf010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/20/2024] [Accepted: 01/08/2025] [Indexed: 01/12/2025]
Abstract
CONTEXT The growth hormone (GH) secretagogue receptor, encoded by GHSR, is expressed on somatotrophs of the pituitary gland. Stimulation with its ligand ghrelin, as well as its constitutive activity, enhances GH secretion. Studies in knockout mice suggest that heterozygous loss-of-function of GHSR is associated with decreased GH response to fasting, but patient observations in small case reports have been equivocal. OBJECTIVE This work aims to establish the phenotype of GHSR haploinsufficiency and its growth response to GH treatment. METHODS This case series includes 26 patients with short stature and heterozygous GHSR variants. Pathogenicity was studied in vitro using total protein levels, cell surface expression, and receptor activity in basal, stimulated, and inhibited states. RESULTS Ten different variants were identified, of which 6 were novel. Variants showed either partial or complete loss of function, primarily through loss of constitutive activity. Patients (aged 4.0-15.1 years) had proportionate short stature (height -2.8 ± 0.5 SDS), failure to thrive with low appetite (n = 4), a mean serum insulin-like growth factor-I (IGF-I) of -1.6 ± 0.7 SDS, and a normal stimulated GH response. Nine patients received GH treatment, showing a height gain of 0.9 ± 0.4 SDS after 1 year and 1.5 ± 0.4 SDS after 2 years (n = 5). CONCLUSION This study combines phenotypical and functional data in a uniquely large group of children with short stature carrying GHSR variants, and shows their good response to GH treatment. The results strengthen the hypothesis of GHSR's role in GH secretion.
Collapse
Affiliation(s)
- Lauren D Punt
- Division of Pediatric Endocrinology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| | - Sander Kooijman
- Division of Endocrinology, Department of Medicine, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| | - Noa J M Mutsters
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, the Netherlands
| | - Kaiming Yue
- Division of Endocrinology, Department of Medicine, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| | - Daniëlle C M van der Kaay
- Division of Pediatric Endocrinology, Department of Pediatrics, Erasmus University Medical Centre, Sophia Children's Hospital, 3015 GD Rotterdam, the Netherlands
| | - Vera van Tellingen
- Department of Pediatrics, Catharina Hospital, 5623 EJ Eindhoven, the Netherlands
| | - Willie M Bakker-van Waarde
- Division of Pediatric Endocrinology, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| | - Annemiek M Boot
- Division of Pediatric Endocrinology, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| | - Erica L T van den Akker
- Division of Pediatric Endocrinology, Department of Pediatrics, Erasmus University Medical Centre, Sophia Children's Hospital, 3015 GD Rotterdam, the Netherlands
| | | | - Kirsten de Groote
- Division of Pediatric Endocrinology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| | - Anne R Kruijsen
- Division of Pediatric Endocrinology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| | | | - M Claire Woltering
- Department of Pediatrics, Reinier de Graaf Gasthuis, 2625 AD Delft, the Netherlands
| | - Malou Heijligers
- Department of Clinical Genetics, Maastricht University Medical Centre, 6229 HX Maastricht, the Netherlands
| | - Josine C van der Heyden
- Department of Pediatrics, Franciscus Gasthuis & Vlietland, 3045 PM Rotterdam, the Netherlands
| | - Ellen M N Bannink
- Department of Pediatrics, Tergooi MC, 1212 VG Hilversum, the Netherlands
| | - Tuula Rinne
- Department of Human Genetics, Radboud UMC, 6525 GA Nijmegen, the Netherlands
| | - Sabine E Hannema
- Department of Pediatric Endocrinology, Amsterdam UMC, location Vrije Universiteit, 1081 HV Amsterdam, the Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, 1081 HV Amsterdam, the Netherlands
- Amsterdam Reproduction and Development, 1105 AZ Amsterdam, the Netherlands
| | - Wouter J de Waal
- Department of Pediatrics, Diakonessenhuis, 3582 KE Utrecht, the Netherlands
| | - Lucia C Delemarre
- Department of Pediatrics, Amstelland Hospital, 1186 AM Amstelveen, the Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| | - Christiaan de Bruin
- Division of Pediatric Endocrinology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| | | | - Jenny A Visser
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, the Netherlands
| | - Patric J D Delhanty
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, the Netherlands
| | - Monique Losekoot
- Department of Clinical Genetics, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| | - Jan M Wit
- Division of Pediatric Endocrinology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| | - Sjoerd D Joustra
- Division of Pediatric Endocrinology, Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, 2333 ZA Leiden, the Netherlands
| |
Collapse
|
2
|
McCaw ZR, Dey R, Somineni H, Amar D, Mukherjee S, Sandor K, Karaletsos T, Koller D, Aschard H, Smith GD, MacArthur D, O'Dushlaine C, Soare TW. Pitfalls in performing genome-wide association studies on ratio traits. HGG ADVANCES 2025; 6:100406. [PMID: 39818621 PMCID: PMC11808723 DOI: 10.1016/j.xhgg.2025.100406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/08/2025] [Accepted: 01/08/2025] [Indexed: 01/18/2025] Open
Abstract
Genome-wide association studies (GWASs) are often performed on ratios composed of a numerator trait divided by a denominator trait. Examples include body mass index (BMI) and the waist-to-hip ratio, among many others. Explicitly or implicitly, the goal of forming the ratio is typically to adjust for an association between the numerator and denominator. While forming ratios may be clinically expedient, there are several important issues with performing GWAS on ratios. Forming a ratio does not "adjust" for the denominator in the sense of conditioning on it, and it is unclear whether associations with ratios are attributable to the numerator, the denominator, or both. Here we demonstrate that associations arising in ratio GWAS can be entirely denominator driven, implying that at least some associations uncovered by ratio GWAS may be due solely to a putative adjustment variable. In a survey of 10 common ratio traits, we find that the ratio model disagrees with the adjusted model (performing GWAS on the numerator while conditioning on the denominator) at around 1/3 of loci. Using BMI as an example, we show that variants detected by only the ratio model are more strongly associated with the denominator (height), while variants detected by only the adjusted model are more strongly associated with the numerator (weight). Although the adjusted model provides effect sizes with a clearer interpretation, it is susceptible to collider bias. We propose and validate a simple method of correcting for the genetic component of collider bias via leave-one-chromosome-out polygenic scoring.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hugues Aschard
- Institut Pasteur, Université Paris Cité, Department of Computational Biology, Paris, France
| | | | - Daniel MacArthur
- Centre for Population Genomics, Garvan Institute of Medical Research, Sydney, NSW, Australia; Centre for Population Genomics, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | | | | |
Collapse
|
3
|
Dreisbach C, Barcelona V, Turchioe MR, Bernstein S, Erickson E. Application of Predictive Analytics in Pregnancy, Birth, and Postpartum Nursing Care. MCN Am J Matern Child Nurs 2025; 50:66-77. [PMID: 39724545 DOI: 10.1097/nmc.0000000000001082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
ABSTRACT Predictive analytics has emerged as a promising approach for improving reproductive health care and patient outcomes. During pregnancy and birth, the ability to accurately predict risks and complications could enable earlier interventions and reduce adverse events. However, there are challenges and ethical considerations for implementing predictive models in perinatal care settings. We introduce major concepts in predictive analytics and describe application of predictive modeling to perinatal care topics such as fertility, preeclampsia, labor onset, vaginal birth after cesarean, uterine rupture, induction outcomes, postpartum hemorrhage, and postpartum mood disorders. Although some predictive models have achieved adequate accuracy (AUC 0.7-0.9), most require additional external validation across diverse populations and practice settings. Bias, particularly racial bias, remains a key limitation of current models. Nurses and advanced practice nurses, including nurse practitioners certified registered nurse anesthetists, and nurse-midwives, play a vital role in ensuring high-quality data collection and communicating predictive model outputs to clinicians and users of the health care system. Addressing the ethical challenges and limitations of predictive analytics is imperative to equitably translate these tools to support patient-centered perinatal care.
Collapse
|
4
|
Alshehhi A, Mousa M, Tay GK, Werghi N, AlSafar H. Genome wide association study reveals novel associations with face morphology. PLoS One 2025; 20:e0299660. [PMID: 39928610 PMCID: PMC11809905 DOI: 10.1371/journal.pone.0299660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 02/12/2024] [Indexed: 02/12/2025] Open
Abstract
Genome-wide association studies (GWAS) on the Middle Eastern population, including the United Arab Emirates (UAE), have been relatively limited. The present study aims to investigate genotype-face morphology associations in the UAE population through Genome Wide Association Studies (GWAS). Phenotypic data (44 face measurements) from 172 Emiratis was obtained through three-dimensional (3D) scanning technology and an automatic face landmarking technique. GWAS analysis revealed associations of 19 genetic loci with six face features, 14 of which are novel. The GWAS analysis revealed 11 significant relationships between 44 face parameters and 242 SNPs, exceeding the GWAS significance threshold. These phenotypes were previously associated with body height, craniofacial defects, and facial characters. The most significant associations of these genetic variations were related to six main facial features which were facial convexity, left orbital protrusion, mandibular contour, nasolabial angle D, inferior facial angle B, and inferior facial angle A. To the best of our knowledge, this is the first GWAS study to investigate the association of SNP variations with face morphology in the Middle Eastern population.
Collapse
Affiliation(s)
- Aamer Alshehhi
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Mira Mousa
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Guan K. Tay
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Division of Psychiatry, Medical School, the University of Western Australia, Crawley, Western Australia, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Naoufel Werghi
- Department Electrical Engineering and Computer Science, C2PS, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Habiba AlSafar
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Emirates Bio-Research Center, Ministry of Interior, Abu Dhabi, United Arab Emirates
| |
Collapse
|
5
|
Vanneste M, Hoskens H, Goovaerts S, Matthews H, Devine J, Aponte JD, Cole J, Shriver M, Marazita ML, Weinberg SM, Walsh S, Richmond S, Klein OD, Spritz RA, Peeters H, Hallgrímsson B, Claes P. Syndrome-informed phenotyping identifies a polygenic background for achondroplasia-like facial variation in the general population. Nat Commun 2024; 15:10458. [PMID: 39622794 PMCID: PMC11612227 DOI: 10.1038/s41467-024-54839-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 11/21/2024] [Indexed: 12/06/2024] Open
Abstract
Human craniofacial shape is highly variable yet highly heritable with numerous genetic variants interacting through multiple layers of development. Here, we hypothesize that Mendelian phenotypes represent the extremes of a phenotypic spectrum and, using achondroplasia as an example, we introduce a syndrome-informed phenotyping approach to identify genomic loci associated with achondroplasia-like facial variation in the general population. We compare three-dimensional facial scans from 43 individuals with achondroplasia and 8246 controls to calculate achondroplasia-like facial scores. Multivariate GWAS of the control scores reveals a polygenic basis for facial variation along an achondroplasia-specific shape axis, identifying genes primarily involved in skeletal development. Jointly modeling these genes in two independent control samples, both human and mouse, shows craniofacial effects approximating the characteristic achondroplasia phenotype. These findings suggest that both complex and Mendelian genetic variation act on the same developmentally determined axes of facial variation, providing insights into the genetic intersection of complex traits and Mendelian disorders.
Collapse
Affiliation(s)
| | - Hanne Hoskens
- Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- McCaig Bone and Joint Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Seppe Goovaerts
- Department of Human Genetics, KU Leuven, Leuven, Belgium
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium
| | - Harold Matthews
- Department of Human Genetics, KU Leuven, Leuven, Belgium
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium
| | - Jay Devine
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven, Belgium
| | - Jose D Aponte
- Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- McCaig Bone and Joint Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Joanne Cole
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Mark Shriver
- Department of Anthropology, Pennsylvania State University, State College, PA, USA
| | - Mary L Marazita
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Seth M Weinberg
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Susan Walsh
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Stephen Richmond
- Applied Clinical Research and Public Health, School of Dentistry, Cardiff University, Cardiff, UK
| | - Ophir D Klein
- Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA, USA
| | - Richard A Spritz
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Hilde Peeters
- Department of Human Genetics, KU Leuven, Leuven, Belgium.
| | - Benedikt Hallgrímsson
- Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- McCaig Bone and Joint Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| | - Peter Claes
- Department of Human Genetics, KU Leuven, Leuven, Belgium.
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium.
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven, Belgium.
| |
Collapse
|
6
|
Weber CJ, Weitzel AJ, Liu AY, Gacasan EG, Sah RL, Cooper KL. Cellular and molecular mechanisms that shape the development and evolution of tail vertebral proportion in mice and jerboas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620311. [PMID: 39484405 PMCID: PMC11527341 DOI: 10.1101/2024.10.25.620311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Despite the functional importance of the vertebral skeleton, little is known about how individual vertebrae elongate or achieve disproportionate lengths as in the giraffe neck. Rodent tails are an abundantly diverse and more tractable system to understand mechanisms of vertebral growth and proportion. In many rodents, disproportionately long mid-tail vertebrae form a 'crescendo-decrescendo' of lengths in the tail series. In bipedal jerboas, these vertebrae grow exceptionally long such that the adult tail is 1.5x the length of a mouse tail, relative to body length, with four fewer vertebrae. How do vertebrae with the same regional identity elongate differently from their neighbors to establish and diversify adult proportion? Here, we find that vertebral lengths are largely determined by differences in growth cartilage height and the number of cells progressing through endochondral ossification. Hypertrophic chondrocyte size, a major contributor to differential elongation in mammal limb bones, differs only in the longest jerboa mid-tail vertebrae where they are exceptionally large. To uncover candidate molecular mechanisms of disproportionate vertebral growth, we performed intersectional RNA-Seq of mouse and jerboa tail vertebrae with similar and disproportionate elongation rates. Many regulators of posterior axial identity and endochondral elongation are disproportionately differentially expressed in jerboa vertebrae. Among these, the inhibitory natriuretic peptide receptor C (NPR3) appears in multiple studies of rodent and human skeletal proportion suggesting it refines local growth rates broadly in the skeleton and broadly in mammals. Consistent with this hypothesis, NPR3 loss of function mice have abnormal tail and limb proportions. Therefore, in addition to genetic components of the complex process of vertebral evolution, these studies reveal fundamental mechanisms of skeletal growth and proportion.
Collapse
Affiliation(s)
- Ceri J Weber
- Department of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Alexander J Weitzel
- Department of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Alexander Y Liu
- Department of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Erica G Gacasan
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Robert L Sah
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Kimberly L Cooper
- Department of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| |
Collapse
|
7
|
Wang SA, Lu MH, Lee AT, Chen CY, Lee LW. Performance of Microsoft Azure Kinect DK as a tool for estimating human body segment lengths. Sci Rep 2024; 14:15811. [PMID: 38982221 PMCID: PMC11233572 DOI: 10.1038/s41598-024-66798-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/04/2024] [Indexed: 07/11/2024] Open
Abstract
The Microsoft Kinect depth sensor, with its built-in software that automatically captures joint coordinates without markers, could be a potential tool for ergonomic studies. This study investigates the performance of Kinect in limb segment lengths using dual-energy X-ray absorptiometry (DXA) as a reference. Healthy children and adults (n = 76) were recruited for limb length measurements by Kinect and DXA. The results showed consistent ratios of arm, forearm, thigh, and leg lengths to height, which were 0.16, 0.14, 0.23, and 0.22 respectively, for both age groups and methods. Kinect exhibited perfect correlation among all limb lengths, indicating fixed proportions assumed by its algorithm. Comparing the two methods, there was a strong correlation (R = 0.850-0.985) and good to excellent agreement (ICC = 0.829-0.977), except for the right leg in adults, where agreement was slightly lower but still moderate (ICC = 0.712). The measurement bias between the methods ranged from - 1.455 to 0.536 cm. In conclusion, Kinect yields outcomes similar to DXA, indicating its potential utility as a tool for ergonomic studies. However, the built-in algorithm of Kinect assumes fixed limb proportions for individuals, which may not be ideal for studies focusing on investigating limb discrepancies or anatomical differences.
Collapse
Affiliation(s)
- Shiou-An Wang
- Department of Computer Science and Information Engineering, Hungkuo Delin University of Technology, New Taipei City, 236302, Taiwan
| | - Ming-Hua Lu
- Department of Computer Science and Information Engineering, Hungkuo Delin University of Technology, New Taipei City, 236302, Taiwan
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital, No. 8, Section West, Jiapu Road, Puzi City, Chiayi, 613016, Taiwan
| | - Ai-Teng Lee
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital, No. 8, Section West, Jiapu Road, Puzi City, Chiayi, 613016, Taiwan
| | - Chao-Yu Chen
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Chiayi, 613016, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Li-Wen Lee
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital, No. 8, Section West, Jiapu Road, Puzi City, Chiayi, 613016, Taiwan.
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan.
| |
Collapse
|
8
|
Schraiber JG, Edge MD. Heritability within groups is uninformative about differences among groups: Cases from behavioral, evolutionary, and statistical genetics. Proc Natl Acad Sci U S A 2024; 121:e2319496121. [PMID: 38470926 PMCID: PMC10962975 DOI: 10.1073/pnas.2319496121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/13/2024] [Indexed: 03/14/2024] Open
Abstract
Without the ability to control or randomize environments (or genotypes), it is difficult to determine the degree to which observed phenotypic differences between two groups of individuals are due to genetic vs. environmental differences. However, some have suggested that these concerns may be limited to pathological cases, and methods have appeared that seem to give-directly or indirectly-some support to claims that aggregate heritable variation within groups can be related to heritable variation among groups. We consider three families of approaches: the "between-group heritability" sometimes invoked in behavior genetics, the statistic [Formula: see text] used in empirical work in evolutionary quantitative genetics, and methods based on variation in ancestry in an admixed population, used in anthropological and statistical genetics. We take up these examples to show mathematically that information on within-group genetic and phenotypic information in the aggregate cannot separate among-group differences into genetic and environmental components, and we provide simulation results that support our claims. We discuss these results in terms of the long-running debate on this topic.
Collapse
Affiliation(s)
- Joshua G. Schraiber
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA90089-2911
| | - Michael D. Edge
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA90089-2911
| |
Collapse
|
9
|
Schraiber JG, Edge MD. Heritability within groups is uninformative about differences among groups: cases from behavioral, evolutionary, and statistical genetics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.06.565864. [PMID: 37986815 PMCID: PMC10659290 DOI: 10.1101/2023.11.06.565864] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Without the ability to control or randomize environments (or genotypes), it is difficult to determine the degree to which observed phenotypic differences between two groups of individuals are due to genetic vs. environmental differences. However, some have suggested that these concerns may be limited to pathological cases, and methods have appeared that seem to give-directly or indirectly-some support to claims that aggregate heritable variation within groups can be related to heritable variation among groups. We consider three families of approaches: the "between-group heritability" sometimes invoked in behavior genetics, the statistic P S T used in empirical work in evolutionary quantitative genetics, and methods based on variation in ancestry in an admixed population, used in anthropological and statistical genetics. We take up these examples to show mathematically that information on within-group genetic and phenotypic information in the aggregate cannot separate among-group differences into genetic and environmental components, and we provide simulation results that support our claims. We discuss these results in terms of the long-running debate on this topic.
Collapse
Affiliation(s)
- Joshua G. Schraiber
- Department of Quantitative and Computational Biology, University of Southern California
| | - Michael D. Edge
- Department of Quantitative and Computational Biology, University of Southern California
| |
Collapse
|
10
|
Dinh-Xuan AT, Graham BL, Thompson B, Miller MR, Stanojevic S. Reconciling the past and considering the future of pulmonary function test interpretation. Eur Respir J 2024; 63:2302225. [PMID: 38302177 DOI: 10.1183/13993003.02225-2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 12/22/2023] [Indexed: 02/03/2024]
Affiliation(s)
- Anh Tuan Dinh-Xuan
- Cochin Hospital, Department of Respiratory and Sleep Medicine, Paris Cité University, Paris, France
| | - Brian L Graham
- Division of Respirology, Critical Care and Sleep Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Bruce Thompson
- Melbourne School of Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Martin R Miller
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Sanja Stanojevic
- Department of Community Health and Epidemiology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
11
|
Sarver DC, Garcia-Diaz J, Saqib M, Riddle RC, Wong GW. Tmem263 deletion disrupts the GH/IGF-1 axis and causes dwarfism and impairs skeletal acquisition. eLife 2024; 12:RP90949. [PMID: 38241182 PMCID: PMC10945605 DOI: 10.7554/elife.90949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2024] Open
Abstract
Genome-wide association studies (GWAS) have identified a large number of candidate genes believed to affect longitudinal bone growth and bone mass. One of these candidate genes, TMEM263, encodes a poorly characterized plasma membrane protein. Single nucleotide polymorphisms in TMEM263 are associated with bone mineral density in humans and mutations are associated with dwarfism in chicken and severe skeletal dysplasia in at least one human fetus. Whether this genotype-phenotype relationship is causal, however, remains unclear. Here, we determine whether and how TMEM263 is required for postnatal growth. Deletion of the Tmem263 gene in mice causes severe postnatal growth failure, proportional dwarfism, and impaired skeletal acquisition. Mice lacking Tmem263 show no differences in body weight within the first 2 weeks of postnatal life. However, by P21 there is a dramatic growth deficit due to a disrupted growth hormone (GH)/insulin-like growth factor 1 (IGF-1) axis, which is critical for longitudinal bone growth. Tmem263-null mice have low circulating IGF-1 levels and pronounced reductions in bone mass and growth plate length. The low serum IGF-1 in Tmem263-null mice is associated with reduced hepatic GH receptor (GHR) expression and GH-induced JAK2/STAT5 signaling. A deficit in GH signaling dramatically alters GH-regulated genes and feminizes the liver transcriptome of Tmem263-null male mice, with their expression profile resembling wild-type female, hypophysectomized male, and Stat5b-null male mice. Collectively, our data validates the causal role for Tmem263 in regulating postnatal growth and raises the possibility that rare mutations or variants of TMEM263 may potentially cause GH insensitivity and impair linear growth.
Collapse
Affiliation(s)
- Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Jean Garcia-Diaz
- Department of Orthopaedic Surgery, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Orthopaedics, University of Maryland School of MedicineBaltimoreUnited States
- Cell and Molecular Medicine graduate program, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Muzna Saqib
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Ryan C Riddle
- Department of Orthopaedic Surgery, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Orthopaedics, University of Maryland School of MedicineBaltimoreUnited States
- Research and Development Service, Baltimore Veterans Administration Medical CenterBaltimoreUnited States
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
12
|
Vanneste M, Hoskens H, Goovaerts S, Matthews H, Aponte JD, Cole J, Shriver M, Marazita ML, Weinberg SM, Walsh S, Richmond S, Klein OD, Spritz RA, Peeters H, Hallgrímsson B, Claes P. Syndrome-informed phenotyping identifies a polygenic background for achondroplasia-like facial variation in the general population. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.07.570544. [PMID: 38106188 PMCID: PMC10723447 DOI: 10.1101/2023.12.07.570544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Human craniofacial shape is highly variable yet highly heritable with genetic variants interacting through multiple layers of development. Here, we hypothesize that Mendelian phenotypes represent the extremes of a phenotypic spectrum and, using achondroplasia as an example, we introduce a syndrome-informed phenotyping approach to identify genomic loci associated with achondroplasia-like facial variation in the normal population. We compared three-dimensional facial scans from 43 individuals with achondroplasia and 8246 controls to calculate achondroplasia-like facial scores. Multivariate GWAS of the control scores revealed a polygenic basis for normal facial variation along an achondroplasia-specific shape axis, identifying genes primarily involved in skeletal development. Jointly modeling these genes in two independent control samples showed craniofacial effects approximating the characteristic achondroplasia phenotype. These findings suggest that both complex and Mendelian genetic variation act on the same developmentally determined axes of facial variation, providing new insights into the genetic intersection of complex traits and Mendelian disorders.
Collapse
Affiliation(s)
| | - Hanne Hoskens
- Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- McCaig Bone and Joint Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Seppe Goovaerts
- Department of Human Genetics, KU Leuven, Leuven, Belgium
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium
| | - Harold Matthews
- Department of Human Genetics, KU Leuven, Leuven, Belgium
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium
| | - Jose D Aponte
- Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- McCaig Bone and Joint Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Joanne Cole
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Mark Shriver
- Department of Anthropology, Pennsylvania State University, State College, PA, USA
| | - Mary L. Marazita
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Seth M. Weinberg
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Susan Walsh
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Stephen Richmond
- Applied Clinical Research and Public Health, School of Dentistry, Cardiff University, Cardiff, UK
| | - Ophir D Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, CA, 94143, USA
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Richard A Spritz
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Hilde Peeters
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Benedikt Hallgrímsson
- Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- McCaig Bone and Joint Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Peter Claes
- Department of Human Genetics, KU Leuven, Leuven, Belgium
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven, Belgium
| |
Collapse
|
13
|
Laustsen BH, Bønløkke JH, Miller MR. How to account for Inuit ancestry in lung function prediction. Int J Circumpolar Health 2023; 82:2151158. [PMID: 36471626 PMCID: PMC9731580 DOI: 10.1080/22423982.2022.2151158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Rigorous lung function prediction equations for the Inuit are lacking. We used spirometry from 351 Inuit and 29 people of other ancestry obtained during an occupational survey in Greenland to determine how to obtain valid lung function predictions for the Inuit using Global Lung Function Initiative (GLI) equations for Europeans. Standing height for the Inuit was used in the predictions as well as their height modified in line with the known differences in standing to sitting height ratio (SHR) for the Inuit. With recorded height in predicting lung function, mean±SD Inuit z-scores for FVC and FEV1 were significantly higher than predicted (0.81±1.20 and 0.53±1.36, respectively, p<0.0001) which was not true for the non-Inuit participants (-0.01±1.04 and 0.15±1.17, respectively). When using height modified for SHR the mean±SD Inuit z-scores for FVC and FEV1 were no longer significantly different from predicted (0.10±1.10 and -0.12±1.24, respectively). The mean±SD Inuit FEV1/FVC z-scores were not significantly different from the non-Inuit, being respectively -0.45±0.98 and -0.01±1.04. Modified height changed the mean±SD Inuit FEV1/FVC z-scores to -0.39±0.99. Representative lung function predictions from GLI equations can be made for Inuit by using standing height modified for the known differences in SHR between Inuit and those of European ancestry.
Collapse
Affiliation(s)
- Birgitte H Laustsen
- Department of Clinical Medicine, Danish Ramazzini Centre, Aalborg University, Aalborg, Denmark,Institute of Nursing & Health Science, Ilisimatusarfik, University of Greenland, Nuuk, Greenland
| | - Jakob H Bønløkke
- Department of Occupational and Environmental Medicine, Danish Ramazzini Centre, Aalborg University Hospital, Aalborg, Denmark
| | - Martin R Miller
- Institute of Applied Birmingham Health Sciences, University of Birmingham, Birmingham, UK,CONTACT Martin R Miller Institute of Applied Birmingham Health Sciences, University of Birmingham, BirminghamUK
| |
Collapse
|
14
|
Sarver DC, Garcia-Diaz J, Saqib M, Riddle RC, Wong GW. Tmem263 deletion disrupts the GH/IGF-1 axis and causes dwarfism and impairs skeletal acquisition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.02.551694. [PMID: 37577461 PMCID: PMC10418210 DOI: 10.1101/2023.08.02.551694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Genome-wide association studies (GWAS) have identified a large number of candidate genes believed to affect longitudinal bone growth and bone mass. One of these candidate genes, TMEM263, encodes a poorly characterized plasma membrane protein. Single nucleotide polymorphisms in TMEM263 are associated with bone mineral density in humans and mutations are associated with dwarfism in chicken and severe skeletal dysplasia in at least one human fetus. Whether this genotype-phenotype relationship is causal, however, remains unclear. Here, we determine whether and how TMEM263 is required for postnatal growth. Deletion of the Tmem263 gene in mice causes severe postnatal growth failure, proportional dwarfism, and impaired skeletal acquisition. Mice lacking Tmem263 show no differences in body weight within the first two weeks of postnatal life. However, by P21 there is a dramatic growth deficit due to a disrupted GH/IGF-1 axis, which is critical for longitudinal bone growth. Tmem263-null mice have low circulating IGF-1 levels and pronounced reductions in bone mass and growth plate length. The low serum IGF-1 in Tmem263-null mice is associated with reduced hepatic GH receptor (GHR) expression and GH-induced JAK2/STAT5 signaling. A deficit in GH signaling dramatically alters GH-regulated genes and feminizes the liver transcriptome of Tmem263-null male mice, with their expression profile resembling a wild-type female, hypophysectomized male, and Stat5b-null male mice. Collectively, our data validates the causal role for Tmem263 in regulating postnatal growth and raises the possibility that rare mutations or variants of TMEM263 may potentially cause GH insensitivity and impair linear growth.
Collapse
Affiliation(s)
- Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jean Garcia-Diaz
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Cell and Molecular Medicine graduate program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Muzna Saqib
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ryan C Riddle
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Research and Development Service, Baltimore Veterans Administration Medical Center, Baltimore, Maryland, USA
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
15
|
Soto ME, Fuentevilla-Alvarez G, Koretzky SG, Vargas-Alarcón G, Torres-Paz YE, Meza-Toledo SE, Pérez-Torres I, Huesca-Gómez C, Gamboa R. Analysis of GPR126 polymorphisms and their relationship with scoliosis in Marfan syndrome and Marfan-like syndrome in Mexican patients. BIOMOLECULES & BIOMEDICINE 2023; 23:976-983. [PMID: 37270806 PMCID: PMC10655884 DOI: 10.17305/bb.2023.9268] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 05/25/2023] [Accepted: 05/25/2023] [Indexed: 06/06/2023]
Abstract
Marfan syndrome (MFS) is an inherited connective tissue disorder. As the spinal growth depends on delicate balance of forces, conditions that affect musculoskeletal matrix often lead to spinal deformities. A large cross-sectional study revealed a 63% prevalence of scoliosis among patients with MFS. Multi-ethnic genome-wide association studies and analyses of human genetic mutations showed that variations and mutations of G protein-coupled receptor 126 (GPR126)locus are associated with multiple skeletal defects, including shorter stature and adolescent idiopathic scoliosis. The study included 54 patients with MFS and 196 control patients. The DNA was extracted from peripheral blood using the saline expulsion method and single nucleotide polymorphism (SNP) determination was carried out using TaqMan probes. Allelic discrimination was performed by RT-qPCR. Significant differences in genotype frequencies were found for SNP rs6570507 in relation to MFS and sex (recessive model, OR 2.46, 95% CI 1.03 -5.87; P = 0.03) and rs7755109 (overdominant model, OR 0.39, 95% CI 0.16-0.91; P = 0.03). The most significant association was found in SNP rs7755109, where the frequency of genotype AG was significantly different between MFS patients with scoliosis and those without (OR 5.68, 95% CI 1.09-29.48; P=0.04). This study, for the first time, examined the genetic association of SNP GPR126 with the risk of scoliosis in patients with connective tissue diseases. The study revealed that SNP rs7755109 is associated with the presence of scoliosis in Mexican patients with MFS.
Collapse
Affiliation(s)
- Maria Elena Soto
- Department of Immunology, Instituto Nacional de Cardiología Ignacio Chávez, México City, México
- Cardiovascular Line Department in American British Cowdray (ABC) Medical Center, México City, México
| | - Giovanny Fuentevilla-Alvarez
- Department of Physiology, Instituto Nacional de Cardiología Ignacio Chávez, México City, México
- Department of Biochemistry, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), México City, México
| | | | | | | | - Sergio Enrique Meza-Toledo
- Department of Biochemistry, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), México City, México
| | - Israel Pérez-Torres
- Department of Cardiovascular Biomedicine, Instituto Nacional de Cardiología Ignacio Chávez, México City, México
| | - Claudia Huesca-Gómez
- Department of Physiology, Instituto Nacional de Cardiología Ignacio Chávez, México City, México
| | - Ricardo Gamboa
- Department of Physiology, Instituto Nacional de Cardiología Ignacio Chávez, México City, México
| |
Collapse
|
16
|
Kun E, Javan EM, Smith O, Gulamali F, de la Fuente J, Flynn BI, Vajrala K, Trutner Z, Jayakumar P, Tucker-Drob EM, Sohail M, Singh T, Narasimhan VM. The genetic architecture and evolution of the human skeletal form. Science 2023; 381:eadf8009. [PMID: 37471560 PMCID: PMC11075689 DOI: 10.1126/science.adf8009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 06/20/2023] [Indexed: 07/22/2023]
Abstract
The human skeletal form underlies bipedalism, but the genetic basis of skeletal proportions (SPs) is not well characterized. We applied deep-learning models to 31,221 x-rays from the UK Biobank to extract a comprehensive set of SPs, which were associated with 145 independent loci genome-wide. Structural equation modeling suggested that limb proportions exhibited strong genetic sharing but were independent of width and torso proportions. Polygenic score analysis identified specific associations between osteoarthritis and hip and knee SPs. In contrast to other traits, SP loci were enriched in human accelerated regions and in regulatory elements of genes that are differentially expressed between humans and great apes. Combined, our work identifies specific genetic variants that affect the skeletal form and ties a major evolutionary facet of human anatomical change to pathogenesis.
Collapse
Affiliation(s)
- Eucharist Kun
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA
| | - Emily M. Javan
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA
| | - Olivia Smith
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA
| | - Faris Gulamali
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Javier de la Fuente
- Department of Psychology, The University of Texas at Austin, Austin, TX, USA
| | - Brianna I. Flynn
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA
| | - Kushal Vajrala
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA
| | - Zoe Trutner
- Department of Surgery and Perioperative Care, The University of Texas at Austin, Austin, TX, USA
| | - Prakash Jayakumar
- Department of Surgery and Perioperative Care, The University of Texas at Austin, Austin, TX, USA
| | | | - Mashaal Sohail
- Centro de Ciencias Genómicas (CCG), Universidad Nacional Autónoma de México (UNAM), 62209 Cuernavaca, Mexico
| | - Tarjinder Singh
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- The New York Genome Center, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute at Columbia University, New York, NY, USA
| | - Vagheesh M. Narasimhan
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA
- Department of Statistics and Data Science, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
17
|
Bartell E, Lin K, Tsuo K, Gan W, Vedantam S, Cole JB, Baronas JM, Yengo L, Marouli E, Amariuta T, Chen Z, Li L, Renthal NE, Jacobsen CM, Salem RM, Walters RG, Hirschhorn JN. Genetics of skeletal proportions in two different populations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.22.541772. [PMID: 37292977 PMCID: PMC10245876 DOI: 10.1101/2023.05.22.541772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Human height can be divided into sitting height and leg length, reflecting growth of different parts of the skeleton whose relative proportions are captured by the ratio of sitting to total height (as sitting height ratio, SHR). Height is a highly heritable trait, and its genetic basis has been well-studied. However, the genetic determinants of skeletal proportion are much less well-characterized. Expanding substantially on past work, we performed a genome-wide association study (GWAS) of SHR in ∼450,000 individuals with European ancestry and ∼100,000 individuals with East Asian ancestry from the UK and China Kadoorie Biobanks. We identified 565 loci independently associated with SHR, including all genomic regions implicated in prior GWAS in these ancestries. While SHR loci largely overlap height-associated loci (P < 0.001), the fine-mapped SHR signals were often distinct from height. We additionally used fine-mapped signals to identify 36 credible sets with heterogeneous effects across ancestries. Lastly, we used SHR, sitting height, and leg length to identify genetic variation acting on specific body regions rather than on overall human height.
Collapse
|
18
|
Kun E, Javan EM, Smith O, Gulamali F, de la Fuente J, Flynn BI, Vajrala K, Trutner Z, Jayakumar P, Tucker-Drob EM, Sohail M, Singh T, Narasimhan VM. The genetic architecture of the human skeletal form. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.03.521284. [PMID: 36712136 PMCID: PMC9881884 DOI: 10.1101/2023.01.03.521284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The human skeletal form underlies our ability to walk on two legs, but unlike standing height, the genetic basis of limb lengths and skeletal proportions is less well understood. Here we applied a deep learning model to 31,221 whole body dual-energy X-ray absorptiometry (DXA) images from the UK Biobank (UKB) to extract 23 different image-derived phenotypes (IDPs) that include all long bone lengths as well as hip and shoulder width, which we analyzed while controlling for height. All skeletal proportions are highly heritable (∼40-50%), and genome-wide association studies (GWAS) of these traits identified 179 independent loci, of which 102 loci were not associated with height. These loci are enriched in genes regulating skeletal development as well as associated with rare human skeletal diseases and abnormal mouse skeletal phenotypes. Genetic correlation and genomic structural equation modeling indicated that limb proportions exhibited strong genetic sharing but were genetically independent of width and torso proportions. Phenotypic and polygenic risk score analyses identified specific associations between osteoarthritis (OA) of the hip and knee, the leading causes of adult disability in the United States, and skeletal proportions of the corresponding regions. We also found genomic evidence of evolutionary change in arm-to-leg and hip-width proportions in humans consistent with striking anatomical changes in these skeletal proportions in the hominin fossil record. In contrast to cardiovascular, auto-immune, metabolic, and other categories of traits, loci associated with these skeletal proportions are significantly enriched in human accelerated regions (HARs), and regulatory elements of genes differentially expressed through development between humans and the great apes. Taken together, our work validates the use of deep learning models on DXA images to identify novel and specific genetic variants affecting the human skeletal form and ties a major evolutionary facet of human anatomical change to pathogenesis.
Collapse
Affiliation(s)
- Eucharist Kun
- Department of Integrative Biology, The University of Texas at Austin
| | - Emily M Javan
- Department of Integrative Biology, The University of Texas at Austin
| | - Olivia Smith
- Department of Integrative Biology, The University of Texas at Austin
| | | | | | - Brianna I Flynn
- Department of Integrative Biology, The University of Texas at Austin
| | - Kushal Vajrala
- Department of Integrative Biology, The University of Texas at Austin
| | - Zoe Trutner
- Department of Surgery and Perioperative Care, The University of Texas at Austin
| | - Prakash Jayakumar
- Department of Surgery and Perioperative Care, The University of Texas at Austin
| | | | - Mashaal Sohail
- Centro de Ciencias Genómicas (CCG), Universidad Nacional Autónoma de México (UNAM)
| | - Tarjinder Singh
- The Department of Psychiatry at Columbia University Irving Medical Center
- The New York Genome Center
- Mortimer B. Zuckerman Mind Brain Behavior Institute at Columbia University
| | - Vagheesh M Narasimhan
- Department of Integrative Biology, The University of Texas at Austin
- Department of Statistics and Data Science, The University of Texas at Austin
| |
Collapse
|
19
|
Connally NJ, Nazeen S, Lee D, Shi H, Stamatoyannopoulos J, Chun S, Cotsapas C, Cassa CA, Sunyaev SR. The missing link between genetic association and regulatory function. eLife 2022; 11:e74970. [PMID: 36515579 PMCID: PMC9842386 DOI: 10.7554/elife.74970] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/02/2022] [Indexed: 12/15/2022] Open
Abstract
The genetic basis of most traits is highly polygenic and dominated by non-coding alleles. It is widely assumed that such alleles exert small regulatory effects on the expression of cis-linked genes. However, despite the availability of gene expression and epigenomic datasets, few variant-to-gene links have emerged. It is unclear whether these sparse results are due to limitations in available data and methods, or to deficiencies in the underlying assumed model. To better distinguish between these possibilities, we identified 220 gene-trait pairs in which protein-coding variants influence a complex trait or its Mendelian cognate. Despite the presence of expression quantitative trait loci near most GWAS associations, by applying a gene-based approach we found limited evidence that the baseline expression of trait-related genes explains GWAS associations, whether using colocalization methods (8% of genes implicated), transcription-wide association (2% of genes implicated), or a combination of regulatory annotations and distance (4% of genes implicated). These results contradict the hypothesis that most complex trait-associated variants coincide with homeostatic expression QTLs, suggesting that better models are needed. The field must confront this deficit and pursue this 'missing regulation.'
Collapse
Affiliation(s)
- Noah J Connally
- Department of Biomedical Informatics, Harvard Medical SchoolBostonUnited States
- Brigham and Women’s Hospital, Division of Genetics, Harvard Medical SchoolBostonUnited States
- Program in Medical and Population Genetics, Broad Institute of MIT and HarvardCambridgeUnited States
| | - Sumaiya Nazeen
- Department of Biomedical Informatics, Harvard Medical SchoolBostonUnited States
- Brigham and Women’s Hospital, Division of Genetics, Harvard Medical SchoolBostonUnited States
- Brigham and Women’s Hospital, Department of Neurology, Harvard Medical SchoolBostonUnited States
| | - Daniel Lee
- Department of Biomedical Informatics, Harvard Medical SchoolBostonUnited States
- Brigham and Women’s Hospital, Division of Genetics, Harvard Medical SchoolBostonUnited States
- Program in Medical and Population Genetics, Broad Institute of MIT and HarvardCambridgeUnited States
| | - Huwenbo Shi
- Program in Medical and Population Genetics, Broad Institute of MIT and HarvardCambridgeUnited States
- Department of Epidemiology, Harvard T.H. Chan School of Public HealthBostonUnited States
| | | | - Sung Chun
- Division of Pulmonary Medicine, Boston Children’s HospitalBostonUnited States
| | - Chris Cotsapas
- Program in Medical and Population Genetics, Broad Institute of MIT and HarvardCambridgeUnited States
- Department of Neurology, Yale Medical SchoolNew HavenUnited States
- Department of Genetics, Yale Medical SchoolNew HavenUnited States
| | - Christopher A Cassa
- Brigham and Women’s Hospital, Division of Genetics, Harvard Medical SchoolBostonUnited States
- Program in Medical and Population Genetics, Broad Institute of MIT and HarvardCambridgeUnited States
| | - Shamil R Sunyaev
- Department of Biomedical Informatics, Harvard Medical SchoolBostonUnited States
- Brigham and Women’s Hospital, Division of Genetics, Harvard Medical SchoolBostonUnited States
- Program in Medical and Population Genetics, Broad Institute of MIT and HarvardCambridgeUnited States
| |
Collapse
|
20
|
Prioritized candidate causal haplotype blocks in plant genome-wide association studies. PLoS Genet 2022; 18:e1010437. [PMID: 36251695 PMCID: PMC9612827 DOI: 10.1371/journal.pgen.1010437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 10/27/2022] [Accepted: 09/20/2022] [Indexed: 11/05/2022] Open
Abstract
Genome wide association studies (GWAS) can play an essential role in understanding genetic basis of complex traits in plants and animals. Conventional SNP-based linear mixed models (LMM) that marginally test single nucleotide polymorphisms (SNPs) have successfully identified many loci with major and minor effects in many GWAS. In plant, the relatively small population size in GWAS and the high genetic diversity found in many plant species can impede mapping efforts on complex traits. Here we present a novel haplotype-based trait fine-mapping framework, HapFM, to supplement current GWAS methods. HapFM uses genotype data to partition the genome into haplotype blocks, identifies haplotype clusters within each block, and then performs genome-wide haplotype fine-mapping to prioritize the candidate causal haplotype blocks of trait. We benchmarked HapFM, GEMMA, BSLMM, GMMAT, and BLINK in both simulated and real plant GWAS datasets. HapFM consistently resulted in higher mapping power than the other GWAS methods in high polygenicity simulation setting. Moreover, it resulted in smaller mapping intervals, especially in regions of high LD, achieved by prioritizing small candidate causal blocks in the larger haplotype blocks. In the Arabidopsis flowering time (FT10) datasets, HapFM identified four novel loci compared to GEMMA’s results, and the average mapping interval of HapFM was 9.6 times smaller than that of GEMMA. In conclusion, HapFM is tailored for plant GWAS to result in high mapping power on complex traits and improved on mapping resolution to facilitate crop improvement. Genome-wide association studies (GWAS) are commonly used in human and plant studies to identify genetic variants responsible for the phenotype of interest and provide foundations for studying disease mechanisms and crop improvement. Most GWAS models are developed and optimized using human datasets. However, the difference between human and plant datasets essentially limits their applications in plant studies, especially when mapping complex traits such as drought resistance and yield. In this study, we present a novel GWAS method, HapFM, tailored for plant datasets to overcome the difficulties of many conventional GWAS methods. HapFM resulted in higher statistical power than conventional GWAS methods for mapping complex traits in our simulation and real dataset analyses. In addition, HapFM reduced the mapping interval by prioritizing candidate causal regions in the genome, which benefits the downstream experimental studies. Last but not least, HapFM can incorporate biological annotations to increase statistical power further. Overall, HapFM balances statistical power, result interpretability, and downstream experimental verifiability.
Collapse
|
21
|
Your height affects your health: genetic determinants and health-related outcomes in Taiwan. BMC Med 2022; 20:250. [PMID: 35831902 PMCID: PMC9281111 DOI: 10.1186/s12916-022-02450-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/22/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Height is an important anthropometric measurement and is associated with many health-related outcomes. Genome-wide association studies (GWASs) have identified hundreds of genetic loci associated with height, mainly in individuals of European ancestry. METHODS We performed genome-wide association analyses and replicated previously reported GWAS-determined single nucleotide polymorphisms (SNPs) in the Taiwanese Han population (Taiwan Biobank; n = 67,452). A genetic instrument composed of 251 SNPs was selected from our GWAS, based on height and replication results as the best-fit polygenic risk score (PRS), in accordance with the clumping and p-value threshold method. We also examined the association between genetically determined height (PRS251) and measured height (phenotype). We performed observational (phenotype) and genetic PRS251 association analyses of height and health-related outcomes. RESULTS GWAS identified 6843 SNPs in 89 genomic regions with genome-wide significance, including 18 novel loci. These were the most strongly associated genetic loci (EFEMP1, DIS3L2, ZBTB38, LCORL, HMGA1, CS, and GDF5) previously reported to play a role in height. There was a positive association between PRS251 and measured height (p < 0.001). Of the 14 traits and 49 diseases analyzed, we observed significant associations of measured and genetically determined height with only eight traits (p < 0.05/[14 + 49]). Height was positively associated with body weight, waist circumference, and hip circumference but negatively associated with body mass index, waist-hip ratio, body fat, total cholesterol, and low-density lipoprotein cholesterol (p < 0.05/[14 + 49]). CONCLUSIONS This study contributes to the understanding of the genetic features of height and health-related outcomes in individuals of Han Chinese ancestry in Taiwan.
Collapse
|
22
|
Graham BL, Miller MR, Thompson BR. Addressing the effect of ancestry on lung volume. Eur Respir J 2022; 59:59/6/2200882. [PMID: 35714993 DOI: 10.1183/13993003.00882-2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/05/2022]
Affiliation(s)
- Brian L Graham
- Division of Respirology, Critical Care and Sleep Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Martin R Miller
- Institute of Applied Health Sciences, University of Birmingham, Birmingham, UK
| | - Bruce R Thompson
- Melbourne School of Health Science, University of Melbourne, Victoria, Australia
| |
Collapse
|
23
|
The genomic origins of the world's first farmers. Cell 2022; 185:1842-1859.e18. [PMID: 35561686 PMCID: PMC9166250 DOI: 10.1016/j.cell.2022.04.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/04/2022] [Accepted: 04/06/2022] [Indexed: 11/24/2022]
Abstract
The precise genetic origins of the first Neolithic farming populations in Europe and Southwest Asia, as well as the processes and the timing of their differentiation, remain largely unknown. Demogenomic modeling of high-quality ancient genomes reveals that the early farmers of Anatolia and Europe emerged from a multiphase mixing of a Southwest Asian population with a strongly bottlenecked western hunter-gatherer population after the last glacial maximum. Moreover, the ancestors of the first farmers of Europe and Anatolia went through a period of extreme genetic drift during their westward range expansion, contributing highly to their genetic distinctiveness. This modeling elucidates the demographic processes at the root of the Neolithic transition and leads to a spatial interpretation of the population history of Southwest Asia and Europe during the late Pleistocene and early Holocene.
Collapse
|
24
|
Wit JM, Joustra SD, Losekoot M, van Duyvenvoorde HA, de Bruin C. Differential Diagnosis of the Short IGF-I-Deficient Child with Apparently Normal Growth Hormone Secretion. Horm Res Paediatr 2022; 94:81-104. [PMID: 34091447 DOI: 10.1159/000516407] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 04/08/2021] [Indexed: 11/19/2022] Open
Abstract
The current differential diagnosis for a short child with low insulin-like growth factor I (IGF-I) and a normal growth hormone (GH) peak in a GH stimulation test (GHST), after exclusion of acquired causes, includes the following disorders: (1) a decreased spontaneous GH secretion in contrast to a normal stimulated GH peak ("GH neurosecretory dysfunction," GHND) and (2) genetic conditions with a normal GH sensitivity (e.g., pathogenic variants of GH1 or GHSR) and (3) GH insensitivity (GHI). We present a critical appraisal of the concept of GHND and the role of 12- or 24-h GH profiles in the selection of children for GH treatment. The mean 24-h GH concentration in healthy children overlaps with that in those with GH deficiency, indicating that the previously proposed cutoff limit (3.0-3.2 μg/L) is too high. The main advantage of performing a GH profile is that it prevents about 20% of false-positive test results of the GHST, while it also detects a low spontaneous GH secretion in children who would be considered GH sufficient based on a stimulation test. However, due to a considerable burden for patients and the health budget, GH profiles are only used in few centres. Regarding genetic causes, there is good evidence of the existence of Kowarski syndrome (due to GH1 variants) but less on the role of GHSR variants. Several genetic causes of (partial) GHI are known (GHR, STAT5B, STAT3, IGF1, IGFALS defects, and Noonan and 3M syndromes), some responding positively to GH therapy. In the final section, we speculate on hypothetical causes.
Collapse
Affiliation(s)
- Jan M Wit
- Department of Paediatrics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Sjoerd D Joustra
- Department of Paediatrics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Monique Losekoot
- Department of Clinical Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | | | - Christiaan de Bruin
- Department of Paediatrics, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
25
|
Saxena A, Sharma V, Muthuirulan P, Neufeld SJ, Tran MP, Gutierrez HL, Chen KD, Erberich JM, Birmingham A, Capellini TD, Cobb J, Hiller M, Cooper KL. Interspecies transcriptomics identify genes that underlie disproportionate foot growth in jerboas. Curr Biol 2022; 32:289-303.e6. [PMID: 34793695 PMCID: PMC8792248 DOI: 10.1016/j.cub.2021.10.063] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 07/16/2021] [Accepted: 10/28/2021] [Indexed: 01/26/2023]
Abstract
Despite the great diversity of vertebrate limb proportion and our deep understanding of the genetic mechanisms that drive skeletal elongation, little is known about how individual bones reach different lengths in any species. Here, we directly compare the transcriptomes of homologous growth cartilages of the mouse (Mus musculus) and bipedal jerboa (Jaculus jaculus), the latter of which has "mouse-like" arms but extremely long metatarsals of the feet. Intersecting gene-expression differences in metatarsals and forearms of the two species revealed that about 10% of orthologous genes are associated with the disproportionately rapid elongation of neonatal jerboa feet. These include genes and enriched pathways not previously associated with endochondral elongation as well as those that might diversify skeletal proportion in addition to their known requirements for bone growth throughout the skeleton. We also identified transcription regulators that might act as "nodes" for sweeping differences in genome expression between species. Among these, Shox2, which is necessary for proximal limb elongation, has gained expression in jerboa metatarsals where it has not been detected in other vertebrates. We show that Shox2 is sufficient to increase mouse distal limb length, and a nearby putative cis-regulatory region is preferentially accessible in jerboa metatarsals. In addition to mechanisms that might directly promote growth, we found evidence that jerboa foot elongation may occur in part by de-repressing latent growth potential. The genes and pathways that we identified here provide a framework to understand the modular genetic control of skeletal growth and the remarkable malleability of vertebrate limb proportion.
Collapse
Affiliation(s)
- Aditya Saxena
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Virag Sharma
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, Dresden 01307, Germany; Max Planck Institute for the Physics of Complex Systems, Nothnitzerstraße 38, Dresden 01187, Germany
| | - Pushpanathan Muthuirulan
- Department of Human Evolutionary Biology, Harvard University, 11 Divinity Avenue, Cambridge, MA 02138, USA
| | - Stanley J Neufeld
- Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Mai P Tran
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Haydee L Gutierrez
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Kevin D Chen
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Joel M Erberich
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Amanda Birmingham
- Center for Computational Biology and Bioinformatics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Terence D Capellini
- Department of Human Evolutionary Biology, Harvard University, 11 Divinity Avenue, Cambridge, MA 02138, USA
| | - John Cobb
- Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Michael Hiller
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, Dresden 01307, Germany; Max Planck Institute for the Physics of Complex Systems, Nothnitzerstraße 38, Dresden 01187, Germany
| | - Kimberly L Cooper
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
26
|
Mäkitie RE, Toiviainen-Salo S, Kaitila I, Mäkitie O. A Novel Osteochondrodysplasia With Empty Sella Associates With a TBX2 Variant. Front Endocrinol (Lausanne) 2022; 13:845889. [PMID: 35311234 PMCID: PMC8927981 DOI: 10.3389/fendo.2022.845889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Skeletal dysplasias comprise a heterogenous group of developmental disorders of skeletal and cartilaginous tissues. Several different forms have been described and the full spectrum of their clinical manifestations and underlying genetic causes are still incompletely understood. We report a three-generation Finnish family with an unusual, autosomal dominant form of osteochondrodysplasia and an empty sella. Affected individuals (age range 24-44 years) exhibit unusual codfish-shaped vertebrae, severe early-onset and debilitating osteoarthritis and an empty sella without endocrine abnormalities. Clinical characteristics also include mild dysmorphic features, reduced sitting height ratio, and obesity. Whole-exome sequencing excluded known skeletal dysplasias and identified a novel heterozygous missense mutation c.899C>T (p.Thr300Met) in TBX2, confirmed by Sanger sequencing. TBX2 is important for development of the skeleton and the brain and three prior reports have described variations in TBX2 in patients portraying a complex phenotype with vertebral anomalies, craniofacial dysmorphism and endocrine dysfunctions. Our mutation lies near a previously reported disease-causing variant and is predicted pathogenic with deleterious effects on protein function. Our findings expand the current spectrum of skeletal dysplasias, support the association of TBX2 mutations with skeletal dysplasia and suggest a role for TBX2 in development of the spinal and craniofacial structures and the pituitary gland.
Collapse
Affiliation(s)
- Riikka E. Mäkitie
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Otorhinolaryngology–Head and Neck Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- *Correspondence: Riikka E. Mäkitie,
| | - Sanna Toiviainen-Salo
- Children’s Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Medical Imaging Center, Pediatric Radiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Ilkka Kaitila
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland
- Department of Clinical Genetics, Helsinki University Hospital, Helsinki, Finland
| | - Outi Mäkitie
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Children’s Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Molecular Medicine and Surgery and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
27
|
Kouri A, Dandurand RJ, Usmani OS, Chow CW. Exploring the 175-year history of spirometry and the vital lessons it can teach us today. Eur Respir Rev 2021; 30:30/162/210081. [PMID: 34615699 DOI: 10.1183/16000617.0081-2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 07/02/2021] [Indexed: 12/25/2022] Open
Abstract
175 years have elapsed since John Hutchinson introduced the world to his version of an apparatus that had been in development for nearly two centuries, the spirometer. Though he was not the first to build a device that sought to measure breathing and quantify the impact of disease and occupation on lung function, Hutchison coined the terms spirometer and vital capacity that are still in use today, securing his place in medical history. As Hutchinson envisioned, spirometry would become crucial to our growing knowledge of respiratory pathophysiology, from Tiffeneau and Pinelli's work on forced expiratory volumes, to Fry and Hyatt's description of the flow-volume curve. In the 20th century, standardization of spirometry further broadened its reach and prognostic potential. Today, spirometry is recognized as essential to respiratory disease diagnosis, management and research. However, controversy exists in some of its applications, uptake in primary care remains sub-optimal and there are concerns related to the way in which race is factored into interpretation. Moving forward, these failings must be addressed, and innovations like Internet-enabled portable spirometers may present novel opportunities. We must also consider the physiologic and practical limitations inherent to spirometry and further investigate complementary technologies such as respiratory oscillometry and other emerging technologies that assess lung function. Through an exploration of the storied history of spirometry, we can better contextualize its current landscape and appreciate the trends that have repeatedly arisen over time. This may help to improve our current use of spirometry and may allow us to anticipate the obstacles confronting emerging pulmonary function technologies.
Collapse
Affiliation(s)
- Andrew Kouri
- Division of Respirology, Dept of Medicine, St. Michael's Hospital, Unity Health Toronto, Ontario, Canada
| | - Ronald J Dandurand
- Lakeshore General Hospital, Quebec, Canada.,Dept of Medicine, Respiratory Division, McGill University, Montreal, Quebec, Canada.,Montreal Chest Institute, Meakins-Christie Labs and Oscillometry Unit of the Centre for Innovative Medicine, McGill University Health Centre and Research Institute, Montreal, Canada
| | - Omar S Usmani
- National Heart and Lung Institute, Imperial College London and Royal Brompton Hospital, London, UK
| | - Chung-Wai Chow
- Dept of Medicine, University of Toronto, Toronto, Canada.,Division of Respirology and Multi-Organ Transplant Programme, Dept of Medicine, Toronto General Hospital, University Health Network, Toronto, Canada
| |
Collapse
|
28
|
Insight into the Candidate Genes and Enriched Pathways Associated with Height, Length, Length to Height Ratio and Body-Weight of Korean Indigenous Breed, Jindo Dog Using Gene Set Enrichment-Based GWAS Analysis. Animals (Basel) 2021; 11:ani11113136. [PMID: 34827868 PMCID: PMC8614278 DOI: 10.3390/ani11113136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/21/2021] [Accepted: 10/28/2021] [Indexed: 12/14/2022] Open
Abstract
As a companion and hunting dog, height, length, length to height ratio (LHR) and body-weight are the vital economic traits for Jindo dog. Human selection and targeted breeding have produced an extraordinary diversity in these traits. Therefore, the identification of causative markers, genes and pathways that help us to understand the genetic basis of this variability is essential for their selection purposes. Here, we performed a genome-wide association study (GWAS) combined with enrichment analysis on 757 dogs using 118,879 SNPs. The genomic heritability (h2) was 0.33 for height and 0.28 for weight trait in Jindo. At p-value < 5 × 10-5, ten, six, thirteen and eleven SNPs on different chromosomes were significantly associated with height, length, LHR and body-weight traits, respectively. Based on our results, HHIP, LCORL and NCAPG for height, IGFI and FGFR3 for length, DLK1 and EFEMP1 for LHR and PTPN2, IGFI and RASAL2 for weight can be the potential candidate genes because of the significant SNPs located in their intronic or upstream regions. The gene-set enrichment analysis highlighted here nine and seven overlapping significant (p < 0.05) gene ontology (GO) terms and pathways among traits. Interestingly, the highlighted pathways were related to hormone synthesis, secretion and signalling were generally involved in the metabolism, growth and development process. Our data provide an insight into the significant genes and pathways if verified further, which will have a significant effect on the breeding of the Jindo dog's population.
Collapse
|
29
|
Bhakta NR, Kaminsky DA, Bime C, Thakur N, Hall GL, McCormack MC, Stanojevic S. Addressing Race in Pulmonary Function Testing by Aligning Intent and Evidence With Practice and Perception. Chest 2021; 161:288-297. [PMID: 34437887 PMCID: PMC8783030 DOI: 10.1016/j.chest.2021.08.053] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/29/2021] [Accepted: 08/17/2021] [Indexed: 10/27/2022] Open
Abstract
The practice of using race or ethnicity in medicine to explain differences between individuals is being called into question because it may contribute to biased medical care and research that perpetuates health disparities and structural racism. A commonly cited example is the use of race or ethnicity in the interpretation of pulmonary function test (PFT) results, yet the perspectives of practicing pulmonologists and physiologists are missing from this discussion. This discussion has global relevance for increasingly multicultural communities in which the range of values that represent normal lung function is uncertain. We review the underlying sources of differences in lung function, including those that may be captured by race or ethnicity, and demonstrate how the current practice of PFT measurement and interpretation is imperfect in its ability to describe accurately the relationship between function and health outcomes. We summarize the arguments against using race-specific equations as well as address concerns about removing race from the interpretation of PFT results. Further, we outline knowledge gaps and critical questions that need to be answered to change the current approach of including race or ethnicity in PFT results interpretation thoughtfully. Finally, we propose changes in interpretation strategies and future research to reduce health disparities.
Collapse
Affiliation(s)
- Nirav R Bhakta
- University of California, San Francisco, San Francisco, CA.
| | | | - Christian Bime
- College of Medicine, The University of Arizona Health Science, Tucson, AZ
| | - Neeta Thakur
- University of California, San Francisco, San Francisco, CA; Zuckerberg San Francisco General Hospital, San Francisco, CA
| | - Graham L Hall
- Children's Lung Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute and School of Allied Health, Curtin University, Perth, WA, Australia
| | | | | |
Collapse
|
30
|
Livingstone KM, Tan MH, Abbott G, Duckham RL, Croft L, Ward J, McEvoy M, Keske MA, Austin C, Bowe SJ. Discovery Genome-Wide Association Study of Body Composition in 4,386 Adults From the UK Biobank's Pilot Imaging Enhancement Study. Front Endocrinol (Lausanne) 2021; 12:692677. [PMID: 34239500 PMCID: PMC8259458 DOI: 10.3389/fendo.2021.692677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/03/2021] [Indexed: 11/24/2022] Open
Abstract
Body composition (fat, skeletal muscle and bone mass) is an important determinant of overall health and risk of endocrine disorders such as type 2 diabetes and osteoporosis. Although diet and physical activity are strongly implicated, body composition is also heritable. We conducted a discovery genome-wide association study on 31 phenotypes from the three-compartment body composition model (fat, lean and bone mass) in a set of 4 386 individuals (n = 2 109 males, n = 2 294 females) from the UK Biobank pilot imaging enhancement program that underwent a dual energy X-ray absorptiometry (DXA) scan for assessment of body composition and genetic screening. From 6 137 607 imputed single nucleotide polymorphisms (SNPs) we identified 17 body composition loci (P<5.0 x 10-8). GWAS from the combined dataset identified four statistically significant SNPs (rs7592270, rs145972737, rs13212044, rs77772562). In sex-stratified GWAS, 10 male specific SNPs across all traits were identified and five female specific SNPs. Of the 17 SNPs, six were in or close to a gene where there was a plausible functional connection. Three SNPs (rs7592270, rs77772562 and rs7552312) were correlated with obesity phenotypes, one SNP (rs2236705) with lean phenotypes and two with bone mass phenotypes (rs112098641 and rs113380185). These results highlight candidate genes and biological pathways related to body composition, including glucose metabolism and estrogen regulation, that are of interest to replicate in future studies.
Collapse
Affiliation(s)
- Katherine M. Livingstone
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, Australia
| | - Mun Hua Tan
- Department of Microbiology and Immunology, Bio21 Institute, University of Melbourne, Melbourne, VIC, Australia
| | - Gavin Abbott
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, Australia
| | - Rachel L. Duckham
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, St Albans, VIC, Australia
| | - Larry Croft
- School of Life and Environmental Sciences, Deakin Genomics Centre, Deakin University, Geelong, VIC, Australia
| | - Joey Ward
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, United Kingdom
| | - Mark McEvoy
- La Trobe Rural Health School, College of Science, Health and Engineering, La Trobe University, Bendigo, VIC, Australia
| | - Michelle A. Keske
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, Australia
| | - Christopher Austin
- School of Life and Environmental Sciences, Deakin Genomics Centre, Deakin University, Geelong, VIC, Australia
| | - Steven J. Bowe
- Deakin Biostatistics Unit, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
31
|
Vicuña L, Norambuena T, Miranda JP, Pereira A, Mericq V, Ongaro L, Montinaro F, Santos JL, Eyheramendy S. Novel loci and Mapuche genetic ancestry are associated with pubertal growth traits in Chilean boys. Hum Genet 2021; 140:1651-1661. [PMID: 34047840 PMCID: PMC8553699 DOI: 10.1007/s00439-021-02290-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 05/04/2021] [Indexed: 01/24/2023]
Abstract
Puberty is a complex developmental process that varies considerably among individuals and populations. Genetic factors explain a large proportion of the variability of several pubertal traits. Recent genome-wide association studies (GWAS) have identified hundreds of variants involved in traits that result from body growth, like adult height. However, they do not capture many genetic loci involved in growth changes over distinct growth phases. Further, such GWAS have been mostly performed in Europeans, but it is unknown how these findings relate to other continental populations. In this study, we analyzed the genetic basis of three pubertal traits; namely, peak height velocity (PV), age at PV (APV) and height at APV (HAPV). We analyzed a cohort of 904 admixed Chilean children and adolescents with European and Mapuche Native American ancestries. Height was measured on roughly a \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$6-$$\end{document}6-month basis from childhood to adolescence between 2006 and 2019. We predict that, in average, HAPV is 4.3 cm higher in European than in Mapuche adolescents (P = 0.042), and APV is 0.73 years later in European compared with Mapuche adolescents (P = 0.023). Further, by performing a GWAS on 774, 433 single-nucleotide polymorphisms, we identified a genetic signal harboring 3 linked variants significantly associated with PV in boys (P\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$< 5 \times 10^{-8}$$\end{document}<5×10-8). This signal has never been associated with growth-related traits.
Collapse
Affiliation(s)
- Lucas Vicuña
- Faculty of Engineering and Sciences, Universidad Adolfo Ibáñez, Peñalolén, Santiago, Chile.,Instituto Milenio de Investigación Sobre los Fundamentos de los Datos (IMFD), Santiago, Chile
| | - Tomás Norambuena
- Faculty of Engineering and Sciences, Universidad Adolfo Ibáñez, Peñalolén, Santiago, Chile.,Instituto Milenio de Investigación Sobre los Fundamentos de los Datos (IMFD), Santiago, Chile
| | - José Patricio Miranda
- Faculty of Engineering and Sciences, Universidad Adolfo Ibáñez, Peñalolén, Santiago, Chile
| | - Ana Pereira
- Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| | - Veronica Mericq
- Institute of Maternal and Child Research, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Linda Ongaro
- Estonian Biocentre, Institute of Genomics, University of Tartu, Riia 23b, 51010, Tartu, Estonia
| | - Francesco Montinaro
- Estonian Biocentre, Institute of Genomics, University of Tartu, Riia 23b, 51010, Tartu, Estonia
| | - José L Santos
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susana Eyheramendy
- Faculty of Engineering and Sciences, Universidad Adolfo Ibáñez, Peñalolén, Santiago, Chile. .,Instituto Milenio de Investigación Sobre los Fundamentos de los Datos (IMFD), Santiago, Chile.
| |
Collapse
|
32
|
Saco-Ledo G, Porta J, Monson TA, Brasil MF, Duyar I. Body proportions according to stature groups in elite athletes. Res Sports Med 2021; 30:516-528. [PMID: 33906546 DOI: 10.1080/15438627.2021.1917402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The aim of this study was to investigate whether body proportions change as stature increases in elite Spanish athletes. The sample includes a total of 2,030 participants, comprised of 1,357 adult males, and 673 adult females. The male athletes were classified into five groups by stature, and the female athletes were classified separately into four stature groups. Ten anthropometric measurements were collected, and eleven body proportions were calculated. The body proportions with significant differences between stature groups in males were relative arm length (0.53-1.60%), relative forearm length (0.69-2.08%), relative thigh length (1.17-1.56%), relative tibial length (1.37-6.39%), cormic index (-0.94 - -4.49%), Manouvrier index (1.60-9.60%), and crural index (1.05-4.79%). In females, the body proportions with significant differences were relative forearm length (1.43%), relative thigh length (1.94-3.88%), relative tibial length (2.74-4.56%), cormic index (-0.74 - -3.72%), and Manouvrier index (1.97-8.71%). The distal parts of the upper and lower limbs increase proportionally as stature increases, whereas relative hand and foot lengths, which are the most distal parts of the extremities, remain constant in elite athletes.
Collapse
Affiliation(s)
- Gonzalo Saco-Ledo
- Catalan School of Kinanthropometry, National Institute of Physical Education, University of Barcelona,Barcelona, Spain.,Bioenergy and Motion Analysis Laboratory, National Research Center on Human Evolution (CENIEH), Burgos, Spain
| | - Jordi Porta
- Catalan School of Kinanthropometry, National Institute of Physical Education, University of Barcelona,Barcelona, Spain
| | - Tesla A Monson
- Department of Anthropology, Western Washington University, Bellingham, USA
| | - Marianne F Brasil
- Human Evolution Research Center, University of California Berkeley, Berkeley, California, USA.,Berkeley Geochronology Center, Berkeley, California, USA
| | - Izzet Duyar
- Department of Anthropology, Faculty of Letters, Istanbul University, Istanbul, Turkey
| |
Collapse
|
33
|
Bartell E, Fujimoto M, Khoury JC, Khoury PR, Vedantam S, Astley CM, Hirschhorn JN, Dauber A. Protein QTL analysis of IGF-I and its binding proteins provides insights into growth biology. Hum Mol Genet 2020; 29:2625-2636. [PMID: 32484228 PMCID: PMC7471503 DOI: 10.1093/hmg/ddaa103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/21/2020] [Accepted: 05/27/2020] [Indexed: 12/15/2022] Open
Abstract
The growth hormone and insulin-like growth factor (IGF) system is integral to human growth. Genome-wide association studies (GWAS) have identified variants associated with height and located near the genes in this pathway. However, mechanisms underlying these genetic associations are not understood. To investigate the regulation of the genes in this pathway and mechanisms by which regulation could affect growth, we performed GWAS of measured serum protein levels of IGF-I, IGF binding protein-3 (IGFBP-3), pregnancy-associated plasma protein A (PAPP-A2), IGF-II and IGFBP-5 in 838 children (3-18 years) from the Cincinnati Genomic Control Cohort. We identified variants associated with protein levels near IGFBP3 and IGFBP5 genes, which contain multiple signals of association with height and other skeletal growth phenotypes. Surprisingly, variants that associate with protein levels at these two loci do not colocalize with height associations, confirmed through conditional analysis. Rather, the IGFBP3 signal (associated with total IGFBP-3 and IGF-II levels) colocalizes with an association with sitting height ratio (SHR); the IGFBP5 signal (associated with IGFBP-5 levels) colocalizes with birth weight. Indeed, height-associated single nucleotide polymorphisms near genes encoding other proteins in this pathway are not associated with serum levels, possibly excluding PAPP-A2. Mendelian randomization supports a stronger causal relationship of measured serum levels with SHR (for IGFBP-3) and birth weight (for IGFBP-5) than with height. In conclusion, we begin to characterize the genetic regulation of serum levels of IGF-related proteins in childhood. Furthermore, our data strongly suggest the existence of growth-regulating mechanisms acting through IGF-related genes in ways that are not reflected in measured serum levels of the corresponding proteins.
Collapse
Affiliation(s)
- Eric Bartell
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Masanobu Fujimoto
- Division of Endocrinology, Cincinnati Center for Growth Disorders, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
- Division of Pediatrics and Perinatology, Tottori University Faculty of Medicine, Yonago, Tottori 683-8504, Japan
| | - Jane C Khoury
- Division of Endocrinology, Cincinnati Center for Growth Disorders, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
- Division of Biostatistics and Epidemiology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Philip R Khoury
- Heart Institute Research Core, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Sailaja Vedantam
- Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Christina M Astley
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Joel N Hirschhorn
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Andrew Dauber
- Division of Endocrinology, Children’s National Hospital, Washington, DC 20010, USA
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| |
Collapse
|
34
|
Lin YJ, Cheng CF, Wang CH, Liang WM, Tang CH, Tsai LP, Chen CH, Wu JY, Hsieh AR, Lee MTM, Lin TH, Liao CC, Huang SM, Zhang Y, Tsai CH, Tsai FJ. Genetic Architecture Associated With Familial Short Stature. J Clin Endocrinol Metab 2020; 105:5805154. [PMID: 32170311 DOI: 10.1210/clinem/dgaa131] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 03/10/2020] [Indexed: 12/21/2022]
Abstract
CONTEXT Human height is an inheritable, polygenic trait under complex and multilocus genetic regulation. Familial short stature (FSS; also called genetic short stature) is the most common type of short stature and is insufficiently known. OBJECTIVE To investigate the FSS genetic profile and develop a polygenic risk predisposition score for FSS risk prediction. DESIGN AND SETTING The FSS participant group of Han Chinese ancestry was diagnosed by pediatric endocrinologists in Taiwan. PATIENTS AND INTERVENTIONS The genetic profiles of 1163 participants with FSS were identified by using a bootstrapping subsampling and genome-wide association studies (GWAS) method. MAIN OUTCOME MEASURES Genetic profile, polygenic risk predisposition score for risk prediction. RESULTS Ten novel genetic single nucleotide polymorphisms (SNPs) and 9 reported GWAS human height-related SNPs were identified for FSS risk. These 10 novel SNPs served as a polygenic risk predisposition score for FSS risk prediction (area under the curve: 0.940 in the testing group). This FSS polygenic risk predisposition score was also associated with the height reduction regression tendency in the general population. CONCLUSION A polygenic risk predisposition score composed of 10 genetic SNPs is useful for FSS risk prediction and the height reduction tendency. Thus, it might contribute to FSS risk in the Han Chinese population from Taiwan.
Collapse
Affiliation(s)
- Ying-Ju Lin
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chi-Fung Cheng
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- Department of Health Services Administration, China Medical University, Taichung, Taiwan
| | - Chung-Hsing Wang
- Children's Hospital of China Medical University, Taichung, Taiwan
| | - Wen-Miin Liang
- Department of Health Services Administration, China Medical University, Taichung, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Li-Ping Tsai
- Department of Pediatrics, Taipei Tzu Chi Hospital, New Taipei City, Taiwan
| | - Chien-Hsiun Chen
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jer-Yuarn Wu
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ai-Ru Hsieh
- Department of Statistics, Tamkang University, New Taipei City, Taiwan
| | | | - Ting-Hsu Lin
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Chiu-Chu Liao
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Shao-Mei Huang
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Yanfei Zhang
- Genomic Medicine Institute, Geisinger, Danville, Pennsylvania, USA
| | - Chang-Hai Tsai
- Department of Biotechnology and Bioinformatics, Asia University, Taichung, Taiwan
| | - Fuu-Jen Tsai
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Children's Hospital of China Medical University, Taichung, Taiwan
- Department of Biotechnology and Bioinformatics, Asia University, Taichung, Taiwan
| |
Collapse
|
35
|
Salek Ardestani S, Aminafshar M, Zandi Baghche Maryam MB, Banabazi MH, Sargolzaei M, Miar Y. Signatures of selection analysis using whole-genome sequence data reveals novel candidate genes for pony and light horse types. Genome 2020; 63:387-396. [PMID: 32407640 DOI: 10.1139/gen-2020-0001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Natural selection and domestication have shaped modern horse populations, resulting in a vast range of phenotypically diverse breeds. Horse breeds are classified into three types (pony, light, and draft) generally based on their body type. Understanding the genetic basis of horse type variation and selective pressures related to the evolutionary trend can be particularly important for current selection strategies. Whole-genome sequences were generated for 14 pony and 32 light horses to investigate the genetic signatures of selection of the horse type in pony and light horses. In the overlapping extremes of the fixation index and nucleotide diversity results, we found novel genomic signatures of selective sweeps near key genes previously implicated in body measurements including C4ORF33, CRB1, CPN1, FAM13A, and FGF12 that may influence variation in pony and light horse types. This study contributes to a better understanding of the genetic background of differences between pony and light horse types.
Collapse
Affiliation(s)
- Siavash Salek Ardestani
- Department of Animal Science, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran
| | - Mehdi Aminafshar
- Department of Animal Science, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran
| | | | - Mohammad Hossein Banabazi
- Department of Biotechnology, Animal Science Research Institute of Iran, Agricultural Research, Education & Extension Organization, Karaj 3146618361, Iran
| | - Mehdi Sargolzaei
- Department of Pathobiology, University of Guelph, Guelph, ON NIG 2W1, Canada.,Select Sires Inc., Plain City, OH 43064, USA
| | - Younes Miar
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS B2N 5E3, Canada
| |
Collapse
|
36
|
Cooper KL. Developmental and Evolutionary Allometry of the Mammalian Limb Skeleton. Integr Comp Biol 2020; 59:1356-1368. [PMID: 31180500 DOI: 10.1093/icb/icz082] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The variety of limb skeletal proportions enables a remarkable diversity of behaviors that include powered flight in bats and flipper-propelled swimming in whales using extremes of a range of homologous limb architectures. Even within human limbs, bone lengths span more than an order of magnitude from the short finger and toe bones to the long arm and leg bones. Yet all of this diversity arises from embryonic skeletal elements that are each a very similar size at formation. In this review article, I survey what is and is not yet known of the development and evolution of skeletal proportion at multiple hierarchical levels of biological organization. These include the cellular parameters of skeletal elongation in the cartilage growth plate, genes associated with differential growth, and putative gene regulatory mechanisms that would allow both covariant and independent evolution of the forelimbs and hindlimbs and of individual limb segments. Although the genetic mechanisms that shape skeletal proportion are still largely unknown, and most of what is known is limited to mammals, it is becoming increasingly apparent that the diversity of bone lengths is an emergent property of a complex system that controls elongation of individual skeletal elements using a genetic toolkit shared by all.
Collapse
Affiliation(s)
- Kimberly L Cooper
- Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093-0377, USA
| |
Collapse
|
37
|
Abstract
Humans show marked variation in body size around the world, both within and among populations. At present, the tallest people in the world are from the Netherlands and the Balkan countries, while the shortest populations are central African Pygmies. There are genetic, genetic plasticity, developmental, and environmental bases for size variation in Homo sapiens from the recent past and the present. Early populations of Homo species also have shown considerable size variation. Populations from the present and the past are also marked by sexual dimorphism, which, itself, shows group variation. There is abundant evidence for the effects of limited food and disease on human growth and resultant adult body size. This environmental influence has been reflected in "secular trends" (over a span of years) in growth and adult size from socioeconomic prosperity or poverty (availability of resources). Selective and evolutionary advantages of small or large body size also have been documented. Heritability for human height is relatively great with current genome-wide association studies (GWAS) identifying hundreds of genes leading to causes of growth and adult size variation. There are also endocrinological pathways limiting growth. An example is the reduced tissue sensitivity to human growth hormone (HGH) and insulin-like growth factor (IGF-1) in Philippine and African hunter-gatherer populations. In several short-statured hunter-gatherer populations (Asian, African, and South American), it has been hypothesized that short life expectancy has selected for early maturity and truncated growth to enhance fertility. Some island populations of humans and other mammals are thought to have been selected for small size because of limited resources, especially protein. The high-protein content of milk as a staple food may contribute to tall stature in East African pastoral peoples. These and other evolutionary questions linked to life history, male competition, reproduction, and mobility are explored in this paper.
Collapse
|
38
|
Abstract
Measurements of prehistoric human skeletal remains provide a record of changes in height and other anthropometric traits over time. Often, these changes are interpreted in terms of plastic developmental response to shifts in diet, climate, or other environmental factors. These changes can also be genetic in origin, but, until recently, it has been impossible to separate the effects of genetics and environment. Here, we use ancient DNA to directly estimate genetic changes in phenotypes and to identify changes driven not by genetics, but by environment. We show that changes over the past 35,000 y are largely predicted by genetics but also identify specific shifts that are more likely to be environmentally driven. The relative contributions of genetics and environment to temporal and geographic variation in human height remain largely unknown. Ancient DNA has identified changes in genetic ancestry over time, but it is not clear whether those changes in ancestry are associated with changes in height. Here, we directly test whether changes over the past 38,000 y in European height predicted using DNA from 1,071 ancient individuals are consistent with changes observed in 1,159 skeletal remains from comparable populations. We show that the observed decrease in height between the Early Upper Paleolithic and the Mesolithic is qualitatively predicted by genetics. Similarly, both skeletal and genetic height remained constant between the Mesolithic and Neolithic and increased between the Neolithic and Bronze Age. Sitting height changes much less than standing height—consistent with genetic predictions—although genetics predicts a small post-Neolithic increase that is not observed in skeletal remains. Geographic variation in stature is also qualitatively consistent with genetic predictions, particularly with respect to latitude. Finally, we hypothesize that an observed decrease in genetic heel bone mineral density in the Neolithic reflects adaptation to the decreased mobility indicated by decreased femoral bending strength. This study provides a model for interpreting phenotypic changes predicted from ancient DNA and demonstrates how they can be combined with phenotypic measurements to understand the relative contribution of genetic and developmentally plastic responses to environmental change.
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW The goal of the review is to provide a comprehensive overview of the current understanding of the mechanisms underlying variation in human stature. RECENT FINDINGS Human height is an anthropometric trait that varies considerably within human populations as well as across the globe. Historically, much research focus was placed on understanding the biology of growth plate chondrocytes and how modifications to core chondrocyte proliferation and differentiation pathways potentially shaped height attainment in normal as well as pathological contexts. Recently, much progress has been made to improve our understanding regarding the mechanisms underlying the normal and pathological range of height variation within as well as between human populations, and today, it is understood to reflect complex interactions among a myriad of genetic, environmental, and evolutionary factors. Indeed, recent improvements in genetics (e.g., GWAS) and breakthroughs in functional genomics (e.g., whole exome sequencing, DNA methylation analysis, ATAC-sequencing, and CRISPR) have shed light on previously unknown pathways/mechanisms governing pathological and common height variation. Additionally, the use of an evolutionary perspective has also revealed important mechanisms that have shaped height variation across the planet. This review provides an overview of the current knowledge of the biological mechanisms underlying height variation by highlighting new research findings on skeletal growth control with an emphasis on previously unknown pathways/mechanisms influencing pathological and common height variation. In this context, this review also discusses how evolutionary forces likely shaped the genomic architecture of height across the globe.
Collapse
Affiliation(s)
| | - Terence D Capellini
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
40
|
Chan CKF, Gulati GS, Sinha R, Tompkins JV, Lopez M, Carter AC, Ransom RC, Reinisch A, Wearda T, Murphy M, Brewer RE, Koepke LS, Marecic O, Manjunath A, Seo EY, Leavitt T, Lu WJ, Nguyen A, Conley SD, Salhotra A, Ambrosi TH, Borrelli MR, Siebel T, Chan K, Schallmoser K, Seita J, Sahoo D, Goodnough H, Bishop J, Gardner M, Majeti R, Wan DC, Goodman S, Weissman IL, Chang HY, Longaker MT. Identification of the Human Skeletal Stem Cell. Cell 2019; 175:43-56.e21. [PMID: 30241615 DOI: 10.1016/j.cell.2018.07.029] [Citation(s) in RCA: 420] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 01/16/2018] [Accepted: 07/20/2018] [Indexed: 12/14/2022]
Abstract
Stem cell regulation and hierarchical organization of human skeletal progenitors remain largely unexplored. Here, we report the isolation of a self-renewing and multipotent human skeletal stem cell (hSSC) that generates progenitors of bone, cartilage, and stroma, but not fat. Self-renewing and multipotent hSSCs are present in fetal and adult bones and can also be derived from BMP2-treated human adipose stroma (B-HAS) and induced pluripotent stem cells (iPSCs). Gene expression analysis of individual hSSCs reveals overall similarity between hSSCs obtained from different sources and partially explains skewed differentiation toward cartilage in fetal and iPSC-derived hSSCs. hSSCs undergo local expansion in response to acute skeletal injury. In addition, hSSC-derived stroma can maintain human hematopoietic stem cells (hHSCs) in serum-free culture conditions. Finally, we combine gene expression and epigenetic data of mouse skeletal stem cells (mSSCs) and hSSCs to identify evolutionarily conserved and divergent pathways driving SSC-mediated skeletogenesis. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Charles K F Chan
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA.
| | - Gunsagar S Gulati
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Rahul Sinha
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | | | - Michael Lopez
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Ava C Carter
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
| | - Ryan C Ransom
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Andreas Reinisch
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Taylor Wearda
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Matthew Murphy
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Rachel E Brewer
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Lauren S Koepke
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Owen Marecic
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Anoop Manjunath
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Eun Young Seo
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Tripp Leavitt
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Wan-Jin Lu
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Allison Nguyen
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Stephanie D Conley
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Ankit Salhotra
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Thomas H Ambrosi
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Mimi R Borrelli
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Taylor Siebel
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Karen Chan
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Katharina Schallmoser
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Jun Seita
- Medical Sciences Innovation Hub Program, RIKEN, Tokyo 103-0027, Japan
| | - Debashis Sahoo
- Departments of Pediatrics and Computer Science and Engineering, University of California San Diego, San Diego, CA 92093, USA
| | - Henry Goodnough
- Department of Orthopedic Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Julius Bishop
- Department of Orthopedic Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Michael Gardner
- Department of Orthopedic Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Ravindra Majeti
- Department of Medicine, Division of Hematology, Stanford Medicine, Stanford, CA 94305, USA
| | - Derrick C Wan
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Stuart Goodman
- Department of Orthopedic Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Irving L Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
| | - Michael T Longaker
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
41
|
Wendt FR, Novroski NM. Identity informative SNP associations in the UK Biobank. Forensic Sci Int Genet 2019; 42:45-48. [PMID: 31226582 DOI: 10.1016/j.fsigen.2019.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 06/01/2019] [Accepted: 06/13/2019] [Indexed: 10/26/2022]
Abstract
Single nucleotide polymorphisms (SNPs) are amenable to genotyping DNA from degraded, inhibited, and/or ancient substrates due to their relatively small amplicon size. Though they have clear advantages over traditional short tandem repeat (STR) typing for specific casework scenarios, the advances in massively parallel sequencing (MPS) have drastically increased the utility of this marker type. The biallelic nature of SNPs makes them individually less informative than STRs due to limited heterozygosity; however, in sufficiently large multiplexes, identity informative SNPs (iiSNPs) may produce combined random match probabilities comparable to STR typing. Multiple MPS library preparation kits now include iiSNPs and similar to STRs, these loci have been rigorously characterized during multiplex development. The relative accessibility of genome-wide association study (GWAS) summary statistics enables re-investigation of forensically relevant targets in high-quality datasets. Here, 4085 GWASs from the UK Biobank European datasets (UKB; 787 ≤ N ≤ 361,194) were mined for iiSNPs typed by the ForenSeq DNA Signature Prep Kit (Verogen). Seven iiSNPs had genome-wide association (p ≤ 5 × 10-8) with 17 phenotypes in UKB Europeans. Most notably, these relationships involve two outwardly visible characteristics: standing height (rs907100; β = 0.011, p = 1.35 × 10-10) and hair/balding patterns (rs2399332; β = -0.009, p = 3.83 × 10-8). The remaining associations involve red blood cell characteristics and measures of lung function. Though these traits are highly polygenic and the individual SNP effects described here have been refuted empirically, we describe the importance and ease of exploring high-quality, freely accessible data to continuously and robustly characterize new and existing forensically relevant loci.
Collapse
Affiliation(s)
- Frank R Wendt
- Department of Psychiatry, Yale School of Medicine and VA CT Healthcare Center, West Haven, CT 06516, USA.
| | - Nicole Mm Novroski
- Forensic Science Program, Department of Anthropology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
| |
Collapse
|
42
|
Styrkarsdottir U, Stefansson OA, Gunnarsdottir K, Thorleifsson G, Lund SH, Stefansdottir L, Juliusson K, Agustsdottir AB, Zink F, Halldorsson GH, Ivarsdottir EV, Benonisdottir S, Jonsson H, Gylfason A, Norland K, Trajanoska K, Boer CG, Southam L, Leung JCS, Tang NLS, Kwok TCY, Lee JSW, Ho SC, Byrjalsen I, Center JR, Lee SH, Koh JM, Lohmander LS, Ho-Pham LT, Nguyen TV, Eisman JA, Woo J, Leung PC, Loughlin J, Zeggini E, Christiansen C, Rivadeneira F, van Meurs J, Uitterlinden AG, Mogensen B, Jonsson H, Ingvarsson T, Sigurdsson G, Benediktsson R, Sulem P, Jonsdottir I, Masson G, Holm H, Norddahl GL, Thorsteinsdottir U, Gudbjartsson DF, Stefansson K. GWAS of bone size yields twelve loci that also affect height, BMD, osteoarthritis or fractures. Nat Commun 2019; 10:2054. [PMID: 31053729 PMCID: PMC6499783 DOI: 10.1038/s41467-019-09860-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 04/03/2019] [Indexed: 12/12/2022] Open
Abstract
Bone area is one measure of bone size that is easily derived from dual-energy X-ray absorptiometry (DXA) scans. In a GWA study of DXA bone area of the hip and lumbar spine (N ≥ 28,954), we find thirteen independent association signals at twelve loci that replicate in samples of European and East Asian descent (N = 13,608 - 21,277). Eight DXA area loci associate with osteoarthritis, including rs143384 in GDF5 and a missense variant in COL11A1 (rs3753841). The strongest DXA area association is with rs11614913[T] in the microRNA MIR196A2 gene that associates with lumbar spine area (P = 2.3 × 10-42, β = -0.090) and confers risk of hip fracture (P = 1.0 × 10-8, OR = 1.11). We demonstrate that the risk allele is less efficient in repressing miR-196a-5p target genes. We also show that the DXA area measure contributes to the risk of hip fracture independent of bone density.
Collapse
Affiliation(s)
| | | | | | | | - Sigrun H Lund
- deCODE genetics/Amgen Inc., Reykjavik, 101, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | | | | | | | - Florian Zink
- deCODE genetics/Amgen Inc., Reykjavik, 101, Iceland
| | | | | | | | | | | | | | - Katerina Trajanoska
- Department of Epidemiology, ErasmusMC, 3015 GD, Rotterdam, The Netherlands
- Department of Internal Medicine, ErasmusMC, 3015 GD, Rotterdam, the Netherlands
| | - Cindy G Boer
- Department of Internal Medicine, ErasmusMC, 3015 GD, Rotterdam, the Netherlands
| | - Lorraine Southam
- Wellcome Trust Sanger Institute, Hinxton, CB10 1SA, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Jason C S Leung
- Jockey Club Centre for Osteoporosis Care and Control, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Nelson L S Tang
- Faculty of Medicine, Department of Chemical Pathology and Laboratory for Genetics of Disease Susceptibility, Li Ka Shing Institute of Health Sciences,, The Chinese University of Hong Kong, Hong Kong, China
- CUHK Shenzhen Research Institute, Shenzhen, 518000, China
| | - Timothy C Y Kwok
- Jockey Club Centre for Osteoporosis Care and Control, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Hong Kong, China
| | - Jenny S W Lee
- Faculty of Medicine, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Department of Medicine, Alice Ho Miu Ling Nethersole Hospital and Tai Po Hospital, Hong Kong, China
| | - Suzanne C Ho
- JC School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | | | - Jacqueline R Center
- Bone Biology Division, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
- School of Medicine Sydney, University of Notre Dame Australia, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, 2010, Australia
| | - Seung Hun Lee
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Jung-Min Koh
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - L Stefan Lohmander
- Orthopaedics, Department of Clinical Sciences Lund, Lund University, SE-22 100, Lund, Sweden
| | - Lan T Ho-Pham
- Bone and Muscle Research Lab, Ton Duc Thang University, Ho Chi Minh City, 700000, Vietnam
| | - Tuan V Nguyen
- Bone Biology Division, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, 2010, Australia
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - John A Eisman
- Bone Biology Division, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
- School of Medicine Sydney, University of Notre Dame Australia, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, 2010, Australia
- Clinical Translation and Advanced Education, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
| | - Jean Woo
- Faculty of Medicine, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Ping-C Leung
- Jockey Club Centre for Osteoporosis Care and Control, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - John Loughlin
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, NE1 7RU, UK
| | - Eleftheria Zeggini
- Wellcome Trust Sanger Institute, Hinxton, CB10 1SA, UK
- Institute of Translational Genomics, Helmholtz Zentrum München, 85764, München, Germany
| | | | - Fernando Rivadeneira
- Department of Epidemiology, ErasmusMC, 3015 GD, Rotterdam, The Netherlands
- Department of Internal Medicine, ErasmusMC, 3015 GD, Rotterdam, the Netherlands
| | - Joyce van Meurs
- Department of Internal Medicine, ErasmusMC, 3015 GD, Rotterdam, the Netherlands
| | | | - Brynjolfur Mogensen
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
- Department of Emergengy Medicine, Landspitali, The National University Hospital of Iceland, 101, Reykjavik, Iceland
- Research Institute in Emergency Medicine, Landspitali, The National University Hospital of Iceland, and University of Iceland, 101, Reykjavik, Iceland
| | - Helgi Jonsson
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
- Department of Medicine, Landspitali-The National University Hospital of Iceland, 101, Reykjavik, Iceland
| | - Thorvaldur Ingvarsson
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
- Department of Orthopedic Surgery, Akureyri Hospital, 600, Akureyri, Iceland
- Institution of Health Science, University of Akureyri, 600, Akureyri, Iceland
| | - Gunnar Sigurdsson
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
- Research Service Center, Reykjavik, 201, Iceland
- Department of Endocrinology and Metabolism, Landspitali, The National University Hospital of Iceland, 101, Reykjavik, Iceland
| | - Rafn Benediktsson
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
- Department of Endocrinology and Metabolism, Landspitali, The National University Hospital of Iceland, 101, Reykjavik, Iceland
| | | | - Ingileif Jonsdottir
- deCODE genetics/Amgen Inc., Reykjavik, 101, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
- Department of Immunology, Landspitali-The National University Hospital of Iceland, 101, Reykjavik, Iceland
| | - Gisli Masson
- deCODE genetics/Amgen Inc., Reykjavik, 101, Iceland
| | - Hilma Holm
- deCODE genetics/Amgen Inc., Reykjavik, 101, Iceland
| | | | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen Inc., Reykjavik, 101, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | - Daniel F Gudbjartsson
- deCODE genetics/Amgen Inc., Reykjavik, 101, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, 107, Iceland
| | - Kari Stefansson
- deCODE genetics/Amgen Inc., Reykjavik, 101, Iceland.
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland.
| |
Collapse
|
43
|
Yue S, Whalen P, Jee YH. Genetic regulation of linear growth. Ann Pediatr Endocrinol Metab 2019; 24:2-14. [PMID: 30943674 PMCID: PMC6449614 DOI: 10.6065/apem.2019.24.1.2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 03/07/2019] [Indexed: 12/20/2022] Open
Abstract
Linear growth occurs at the growth plate. Therefore, genetic defects that interfere with the normal function of the growth plate can cause linear growth disorders. Many genetic causes of growth disorders have already been identified in humans. However, recent genome-wide approaches have broadened our knowledge of the mechanisms of linear growth, not only providing novel monogenic causes of growth disorders but also revealing single nucleotide polymorphisms in genes that affect height in the general population. The genes identified as causative of linear growth disorders are heterogeneous, playing a role in various growth-regulating mechanisms including those involving the extracellular matrix, intracellular signaling, paracrine signaling, endocrine signaling, and epigenetic regulation. Understanding the underlying genetic defects in linear growth is important for clinicians and researchers in order to provide proper diagnoses, management, and genetic counseling, as well as to develop better treatment approaches for children with growth disorders.
Collapse
Affiliation(s)
- Shanna Yue
- Pediatric Endocrine, Metabolism and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Philip Whalen
- Pediatric Endocrine, Metabolism and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Youn Hee Jee
- Pediatric Endocrine, Metabolism and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA,Address for correspondence: Youn Hee Jee, MD Pediatric Endocrine, Metabolism and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, CRC, Room 1-3330, 10 Center Drive MSC 1103, Bethesda, MD 20892-1103, USA Tel: +1-301-435-5834 Fax: +1-301-402-0574 E-mail:
| |
Collapse
|
44
|
Koromani F, Trajanoska K, Rivadeneira F, Oei L. Recent Advances in the Genetics of Fractures in Osteoporosis. Front Endocrinol (Lausanne) 2019; 10:337. [PMID: 31231309 PMCID: PMC6559287 DOI: 10.3389/fendo.2019.00337] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 05/10/2019] [Indexed: 12/15/2022] Open
Abstract
Genetic susceptibility, together with old age, female sex, and low bone mineral density (BMD) are amongst the strongest determinants of fracture risk. Tmost recent large-scale genome-wide association study (GWAS) meta-analysis has yielded fifteen loci. This review focuses on the advances in the research of genetic determinants of fracture risk. We first discuss the genetic architecture of fracture risk, touching upon different methods and overall findings. We then discuss in a second paragraph the most recent advances in the field and focus on the genetics of fracture risk and also of other endophenotypes closely related to fracture risk such as bone mineral density (BMD). Application of state-of-the-art methodology such as Mendelian randzation in fracture GWAS are reviewed. The final part of this review touches upon potential future directions in genetic research of osteoporotic fractures.
Collapse
Affiliation(s)
- Fjorda Koromani
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Katerina Trajanoska
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Ling Oei
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
- *Correspondence: Ling Oei
| |
Collapse
|
45
|
Phenotype-Specific Enrichment of Mendelian Disorder Genes near GWAS Regions across 62 Complex Traits. Am J Hum Genet 2018; 103:535-552. [PMID: 30290150 DOI: 10.1016/j.ajhg.2018.08.017] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/28/2018] [Indexed: 01/29/2023] Open
Abstract
Although recent studies provide evidence for a common genetic basis between complex traits and Mendelian disorders, a thorough quantification of their overlap in a phenotype-specific manner remains elusive. Here, we have quantified the overlap of genes identified through large-scale genome-wide association studies (GWASs) for 62 complex traits and diseases with genes containing mutations known to cause 20 broad categories of Mendelian disorders. We identified a significant enrichment of genes linked to phenotypically matched Mendelian disorders in GWAS gene sets; of the total 1,240 comparisons, a higher proportion of phenotypically matched or related pairs (n = 50 of 92 [54%]) than phenotypically unmatched pairs (n = 27 of 1,148 [2%]) demonstrated significant overlap, confirming a phenotype-specific enrichment pattern. Further, we observed elevated GWAS effect sizes near genes linked to phenotypically matched Mendelian disorders. Finally, we report examples of GWAS variants localized at the transcription start site or physically interacting with the promoters of genes linked to phenotypically matched Mendelian disorders. Our results are consistent with the hypothesis that genes that are disrupted in Mendelian disorders are dysregulated by non-coding variants in complex traits and demonstrate how leveraging findings from related Mendelian disorders and functional genomic datasets can prioritize genes that are putatively dysregulated by local and distal non-coding GWAS variants.
Collapse
|
46
|
Guo MH, Hirschhorn JN, Dauber A. Insights and Implications of Genome-Wide Association Studies of Height. J Clin Endocrinol Metab 2018; 103:3155-3168. [PMID: 29982553 PMCID: PMC7263788 DOI: 10.1210/jc.2018-01126] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 06/27/2018] [Indexed: 01/24/2023]
Abstract
CONTEXT In the last decade, genome-wide association studies (GWASs) have catalyzed our understanding of the genetics of height and have identified hundreds of regions of the genome associated with adult height and other height-related body measurements. EVIDENCE ACQUISITION GWASs related to height were identified via PubMed search and a review of the GWAS catalog. EVIDENCE SYNTHESIS The GWAS results demonstrate that height is highly polygenic: that is, many thousands of genetic variants distributed across the genome each contribute to an individual's height. These height-associated regions of the genome are enriched for genes in known biological pathways involved in growth, such as fibroblast growth factor signaling, as well as for genes expressed in relevant tissues, such as the growth plate. GWASs can also uncover previously unappreciated biological pathways, such as the STC2/PAPPA/IGFBP4 pathway. The genes implicated by GWASs are often the same genes that are the genetic causes of Mendelian growth disorders or skeletal dysplasias, and GWAS results can provide complementary information about these disorders. CONCLUSIONS Here, we review the rationale behind GWASs and what we have learned from GWASs for height, including how it has enhanced our understanding of the underlying biology of human growth. We also highlight the implications of GWASs in terms of prediction of adult height and our understanding of Mendelian growth disorders.
Collapse
Affiliation(s)
- Michael H Guo
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- College of Medicine, University of Florida, Gainesville, Florida
| | - Joel N Hirschhorn
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
| | - Andrew Dauber
- Division of Endocrinology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Correspondence and Reprint Requests: Andrew Dauber, MD, MMSc, Division of Endocrinology, Children’s National Medical Center, 111 Michigan Avenue NW, West Wing Floor 3.5, Suite 200, Room 1215, Washington, DC 20010. E-mail:
| |
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW Genome-wide approaches including genome-wide association studies as well as exome and genome sequencing represent powerful new approaches that have improved our ability to identify genetic causes of human disorders. The purpose of this review is to describe recent advances in the genetic causes of short stature. RECENT FINDINGS In addition to SHOX deficiency which is one of the most common causes of isolated short stature, PAPPA2, ACAN, NPPC, NPR2, PTPN11 (and other rasopathies), FBN1, IHH and BMP2 have been identified in isolated growth disorders with or without other mild skeletal findings. In addition, novel genetic causes of syndromic short stature have been discovered, including pathogenic variants in BRCA1, DONSON, AMMECR1, NFIX, SLC25A24, and FN1. SUMMARY Isolated growth disorders are often monogenic. Specific genetic causes typically have specific biochemical and/or phenotype characteristics which are diagnostically helpful. Identification of additional subjects with a specific genetic cause of short stature often leads to a broadening of the known clinical spectrum for that condition. The identification of novel genetic causes of short stature has provided important insights into the underlying molecular mechanisms of growth failure.
Collapse
|
48
|
Perez-Colon S, Lazareva O, Purushothaman R, Malik S, Ten S, Bhangoo A. Baseline IGFBP - 3 as the Key Element to Predict Growth Response to Growth Hormone and IGF - 1 Therapy in Subjects with Non - GH Deficient Short Stature and IGF - 1 Deficiency. Int J Endocrinol Metab 2018; 16:e58928. [PMID: 30197657 PMCID: PMC6113715 DOI: 10.5812/ijem.58928] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 04/05/2018] [Accepted: 04/08/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Short stature in children represents a heterogeneous group with different etiologies. Primary Insulin like growth factor 1 (IGF - 1) deficiency in short stature can present with normal or elevated growth hormone (GH) production. Currently there is no model that can reliably predict response to recombinant (r)GH therapy and/or rIGF - 1 therapy in children with non - GH deficient short stature. HYPOTHESIS Baseline Insulin like growth factor binding protein 3 (IGFBP - 3) along with ∆ IGF - 1 in the first 3 months of GH therapy level can be a marker of growth response to the rGH and/or rIGF - 1 therapy in children with non - growth hormone deficiency short stature. OBJECTIVES To study the relationship between baseline IGFBP - 3 and IGF - 1 levels and the response to rGH and rIGF - 1 therapy in children with short stature, normal GH secretion and low IGF - 1 SDS. METHODS 43 children, age 9.07 ± 2.75 years with height -2.72 ± 0.7 SD and baseline IGF - 1 of -2.76 ± 0.58 SD, who passed the growth hormone releasing hormone (GHRH) stimulation test were included in a retrospective chart review. They were treated with rGH therapy with a mean dose of 0.46 ± 0.1 mg/kg/week. Growth velocity (GV), IGF - 1 and IGFBP - 3 levels were done at 3 and 6 months of therapy. Subjects with poor response to rGH after 6 months of therapy were switched to rIGF - 1 therapy at 0.24 mg/kg/day for the next 6 months. Subjects were divided according to their growth rate into responders to rGH (N = 23); non - responders to rGH, responders to rIGF - 1 (N = 14) and non - responders to rGH and rIGF-1 (N = 6). RESULTS There was no correlation between GV and peak GH level at GHRH test. Growth velocity positively correlated with ΔIGF - 1 SD among subjects treated with rGH therapy. Height SD positively correlated with IGFBP - 3 SD. Baseline IGFBP - 3 also inversely correlated with GH peak during GHRH test. CONCLUSIONS In subjects with short stature and low IGF - 1 level, baseline IGFBP - 3 levels can predict the growth response to rGH and/or rIGF - 1 therapy.
Collapse
Affiliation(s)
- Sheila Perez-Colon
- Division of Pediatric Endocrinology at SUNY Downstate Medical Center and Kings County Hospital, Brooklyn, NY, USA
| | | | | | - Shahid Malik
- Department of Medicine, NYU Woodhull Medical and Mental Health Center Brooklyn, NY, USA
| | | | - Amrit Bhangoo
- Pediatric Endocrinology Children’s Hospital of Orange County, Orange CA, USA
- Corresponding author: Amrit Bhangoo, MD, 1201 W. La Veta Ave., Orange, CA 92868, USA. Tel: +1-7145093364, Fax: +1-7185093300, E-mail:
| |
Collapse
|
49
|
Chan Y, Chan YK, Goodman DB, Guo X, Chavez A, Lim ET, Church GM. Enabling multiplexed testing of pooled donor cells through whole-genome sequencing. Genome Med 2018; 10:31. [PMID: 29673390 PMCID: PMC5909281 DOI: 10.1186/s13073-018-0541-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 04/05/2018] [Indexed: 11/10/2022] Open
Abstract
We describe a method that enables the multiplex screening of a pool of many different donor cell lines. Our method accurately predicts each donor proportion from the pool without requiring the use of unique DNA barcodes as markers of donor identity. Instead, we take advantage of common single nucleotide polymorphisms, whole-genome sequencing, and an algorithm to calculate the proportions from the sequencing data. By testing using simulated and real data, we showed that our method robustly predicts the individual proportions from a mixed-pool of numerous donors, thus enabling the multiplexed testing of diverse donor cells en masse. More information is available at https://pgpresearch.med.harvard.edu/poolseq/
Collapse
Affiliation(s)
- Yingleong Chan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA. .,Department of Genetics, Harvard Medical School, Boston, MA, 02115, USA.
| | - Ying Kai Chan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.,Department of Genetics, Harvard Medical School, Boston, MA, 02115, USA
| | - Daniel B Goodman
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.,Department of Genetics, Harvard Medical School, Boston, MA, 02115, USA.,Harvard-MIT Health Sciences and Technology, Cambridge, MA, 02139, USA
| | - Xiaoge Guo
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.,Department of Genetics, Harvard Medical School, Boston, MA, 02115, USA
| | - Alejandro Chavez
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Elaine T Lim
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.,Department of Genetics, Harvard Medical School, Boston, MA, 02115, USA
| | - George M Church
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA. .,Department of Genetics, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
50
|
Zhang C, Liu J, Iqbal F, Lu Y, Mustafa S, Bukhari F, Lou H, Fu R, Wu Z, Yang X, Bukhari I, Aslam M, Xu S. A missense point mutation in COL10A1 identified with whole-genome deep sequencing in a 7-generation Pakistan dwarf family. Heredity (Edinb) 2017; 120:83-89. [PMID: 29234170 DOI: 10.1038/s41437-017-0021-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 10/10/2017] [Accepted: 10/11/2017] [Indexed: 02/07/2023] Open
Abstract
Disease-associated variants in the human genome are continually being identified using DNA sequencing technologies that are especially effective for Mendelian disorders. Here we sequenced whole genome to high coverage (>30×) of 6 members of a 7-generation family with dwarfism from a consanguineous tribe in Pakistan to determine the causal variant(s). We identified a missense variant rs111033552 (c.2011T>C [p.Ser671Pro]) located in COL10A1 (encodes the alpha chain of type X collagen) as the most likely contributor to the dwarfism. We further confirmed the variant in 22 family members using Sanger sequencing. All affected individuals are heterozygous for the missense mutation rs111033552 and no individual homozygous was observed. Moreover, the mutation was absent in 69,985 individuals representing >150 global populations. Taking advantage of whole-genome sequencing data, we also examined other variant forms, including copy number variation and insertion/deletion, but failed to identify such variants enriched in the affected individuals. Thus rs111033552 had priority for linkage with dwarfism.
Collapse
Affiliation(s)
- Chao Zhang
- Chinese Academy of Sciences (CAS) Key Laboratory of Computational Biology, Max Planck Independent Research Group on Population Genomics, CAS-MPG Partner Institute for Computational Biology (PICB), Shanghai Institutes for Biological Sciences, CAS, Shanghai, 200031, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiaojiao Liu
- Chinese Academy of Sciences (CAS) Key Laboratory of Computational Biology, Max Planck Independent Research Group on Population Genomics, CAS-MPG Partner Institute for Computational Biology (PICB), Shanghai Institutes for Biological Sciences, CAS, Shanghai, 200031, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Furhan Iqbal
- Department of Zoology, Institute of Pure and Applied Biology, Bahauddin Zakariya University Multan, Multan, Pakistan
| | - Yan Lu
- Chinese Academy of Sciences (CAS) Key Laboratory of Computational Biology, Max Planck Independent Research Group on Population Genomics, CAS-MPG Partner Institute for Computational Biology (PICB), Shanghai Institutes for Biological Sciences, CAS, Shanghai, 200031, China
| | - Saima Mustafa
- Department of Zoology, Institute of Pure and Applied Biology, Bahauddin Zakariya University Multan, Multan, Pakistan
| | - Firdous Bukhari
- Department of Zoology, Institute of Pure and Applied Biology, Bahauddin Zakariya University Multan, Multan, Pakistan
| | - Haiyi Lou
- Chinese Academy of Sciences (CAS) Key Laboratory of Computational Biology, Max Planck Independent Research Group on Population Genomics, CAS-MPG Partner Institute for Computational Biology (PICB), Shanghai Institutes for Biological Sciences, CAS, Shanghai, 200031, China
| | - Ruiqing Fu
- Chinese Academy of Sciences (CAS) Key Laboratory of Computational Biology, Max Planck Independent Research Group on Population Genomics, CAS-MPG Partner Institute for Computational Biology (PICB), Shanghai Institutes for Biological Sciences, CAS, Shanghai, 200031, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhendong Wu
- Chinese Academy of Sciences (CAS) Key Laboratory of Computational Biology, Max Planck Independent Research Group on Population Genomics, CAS-MPG Partner Institute for Computational Biology (PICB), Shanghai Institutes for Biological Sciences, CAS, Shanghai, 200031, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Xiong Yang
- Chinese Academy of Sciences (CAS) Key Laboratory of Computational Biology, Max Planck Independent Research Group on Population Genomics, CAS-MPG Partner Institute for Computational Biology (PICB), Shanghai Institutes for Biological Sciences, CAS, Shanghai, 200031, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ihtisham Bukhari
- Department of Biochemistry, Biomarkers Research Program Prince Mutaib Chair for Biomarkers of Osteoporosis, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Muhammad Aslam
- Department of Zoology, Institute of Pure and Applied Biology, Bahauddin Zakariya University Multan, Multan, Pakistan
| | - Shuhua Xu
- Chinese Academy of Sciences (CAS) Key Laboratory of Computational Biology, Max Planck Independent Research Group on Population Genomics, CAS-MPG Partner Institute for Computational Biology (PICB), Shanghai Institutes for Biological Sciences, CAS, Shanghai, 200031, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China. .,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China. .,Collaborative Innovation Center of Genetics and Development, Shanghai, 200438, China.
| |
Collapse
|