Imperial College London

Professor Duncan Bassett

Faculty of MedicineDepartment of Metabolism, Digestion and Reproduction

Professor of Endocrinology
 
 
 
//

Contact

 

+44 (0)20 3313 4613d.bassett Website

 
 
//

Location

 

10N6 Commomwealth BuildingHammersmith HospitalHammersmith Campus

//

Summary

 

Publications

Publication Type
Year
to

135 results found

Loisay L, Komla-Ebri D, Morice A, Heuzé Y, Viaut C, de La Seiglière A, Kaci N, Chan D, Lamouroux A, Baujat G, Bassett JHD, Williams GR, Legeai-Mallet Let al., 2023, Hypochondroplasia gain-of-function mutation in FGFR3 causes defective bone mineralization in mice, JCI Insight, Vol: 8, Pages: 1-21, ISSN: 2379-3708

Hypochondroplasia (HCH) is a mild dwarfism caused by missense mutations in fibroblast growth factor receptor 3 (FGFR3), with the majority of cases resulting from a heterozygous p.Asn540Lys gain-of-function mutation. Here, we report the generation and characterization of the first mouse model (Fgfr3Asn534Lys/+) of HCH to our knowledge. Fgfr3Asn534Lys/+ mice exhibited progressive dwarfism and impairment of the synchondroses of the cranial base, resulting in defective formation of the foramen magnum. The appendicular and axial skeletons were both severely affected and we demonstrated an important role of FGFR3 in regulation of cortical and trabecular bone structure. Trabecular bone mineral density (BMD) of long bones and vertebral bodies was decreased, but cortical BMD increased with age in both tibiae and femurs. These results demonstrate that bones in Fgfr3Asn534Lys/+ mice, due to FGFR3 activation, exhibit some characteristics of osteoporosis. The present findings emphasize the detrimental effect of gain-of-function mutations in the Fgfr3 gene on long bone modeling during both developmental and aging processes, with potential implications for the management of elderly patients with hypochondroplasia and osteoporosis.

Journal article

Kooblall KG, Stevenson M, Stewart M, Harris L, Zalucki O, Dewhurst H, Butterfield N, Leng H, Hough TA, Ma D, Siow B, Potter P, Cox RD, Brown SDM, Horwood N, Wright B, Lockstone H, Buck D, Vincent TL, Hannan FM, Bassett JHD, Williams GR, Lines KE, Piper M, Wells S, Teboul L, Hennekam RC, Thakker RVet al., 2023, A mouse model with a frameshift mutation in the nuclear factor I/X (NFIX) gene has phenotypic features of Marshall-Smith syndrome, JBMR Plus, Vol: 7, ISSN: 2473-4039

The nuclear factor I/X (NFIX) gene encodes a ubiquitously expressed transcription factor whose mutations lead to two allelic disorders characterized by developmental, skeletal, and neural abnormalities, namely, Malan syndrome (MAL) and Marshall–Smith syndrome (MSS). NFIX mutations associated with MAL mainly cluster in exon 2 and are cleared by nonsense-mediated decay (NMD) leading to NFIX haploinsufficiency, whereas NFIX mutations associated with MSS are clustered in exons 6–10 and escape NMD and result in the production of dominant-negative mutant NFIX proteins. Thus, different NFIX mutations have distinct consequences on NFIX expression. To elucidate the in vivo effects of MSS-associated NFIX exon 7 mutations, we used CRISPR-Cas9 to generate mouse models with exon 7 deletions that comprised: a frameshift deletion of two nucleotides (Nfix Del2); in-frame deletion of 24 nucleotides (Nfix Del24); and deletion of 140 nucleotides (Nfix Del140). Nfix+/Del2, Nfix+/Del24, Nfix+/Del140, NfixDel24/Del24, and NfixDel140/Del140 mice were viable, normal, and fertile, with no skeletal abnormalities, but NfixDel2/Del2 mice had significantly reduced viability (p < 0.002) and died at 2–3 weeks of age. Nfix Del2 was not cleared by NMD, and NfixDel2/Del2 mice, when compared to Nfix+/+ and Nfix+/Del2 mice, had: growth retardation; short stature with kyphosis; reduced skull length; marked porosity of the vertebrae with decreased vertebral and femoral bone mineral content; and reduced caudal vertebrae height and femur length. Plasma biochemistry analysis revealed NfixDel2/Del2 mice to have increased total alkaline phosphatase activity but decreased C-terminal telopeptide and procollagen-type-1-N-terminal propeptide concentrations compared to Nfix+/+ and Nfix+/Del2 mice. NfixDel2/Del2 mice were also found to have enlarged cerebral cortices and ventricular areas but smaller dentate gyrus compared to Nfix+/+ mice. Thus, NfixDel2/Del2 mice provide

Journal article

Behmoaras J, 2023, Multinucleation resets human macrophages for specialized functions at the expense of their identity, EMBO Reports, Vol: 24, Pages: 1-19, ISSN: 1469-221X

Macrophages undergo plasma membrane fusion and cell multinucleation to form multinucleated giant cells (MGCs) such as osteoclasts in bone, Langhans giant cells (LGCs) as part of granulomas or foreign-body giant cells (FBGCs) in reaction to exogenous material. How multinucleation per se contributes to functional specialization of mature mononuclear macrophages remains poorly understood in humans. Here, we integrate comparative transcriptomics with functional assays in purified mature mononuclear and multinucleated human osteoclasts, LGCs and FBGCs. Strikingly, in all three types of MGCs, multinucleation causes a pronounced downregulation of macrophage identity. We show enhanced lysosome-mediated intracellular iron homeostasis promoting MGC formation. The transition from mononuclear to multinuclear state is accompanied by cell specialization specific to each polykaryon. Enhanced phagocytic and mitochondrial function associate with FBGCs and osteoclasts, respectively. Moreover, human LGCs preferentially express B7-H3 (CD276) and can form granuloma-like clusters in vitro, suggesting that their multinucleation potentiates T cell activation. These findings demonstrate how cell–cell fusion and multinucleation reset human macrophage identity as part of an advanced maturation step that confers MGC-specific functionality.

Journal article

Loisay L, Ebri DK, Kaci N, Bassett JHD, Williams GR, Legeai-Mallet Let al., 2022, Fgfr3 gain-of-function mutation impacts bone homeostasis in hypochondroplasia mouse model., Annual Meeting of the American-Society-for-Bone-and-Mineral-Research (ASBMR), Publisher: WILEY, Pages: 29-30, ISSN: 0884-0431

Conference paper

Loisay L, Ebri DK, Kaci N, Bassett JHD, Williams GR, Legeai-Mallet Let al., 2022, Fgfr3 gain-of-function mutation impacts bone homeostasis in hypochondroplasia mouse model., Annual Meeting of the American-Society-for-Bone-and-Mineral-Research (ASBMR), Publisher: WILEY, Pages: 250-250, ISSN: 0884-0431

Conference paper

Lademann F, Mayerl S, Tsourdi E, Verrey F, Leitch VD, Williams GR, Bassett JHD, Hofbauer LC, Heuer H, Rauner Met al., 2022, The Thyroid Hormone Transporter MCT10 Is a Novel Regulator of Trabecular Bone Mass and Bone Turnover in Male Mice, ENDOCRINOLOGY, Vol: 163, ISSN: 0013-7227

Journal article

Foessl I, Bassett JHD, Bjornerem A, Busse B, Calado A, Chavassieux P, Christou M, Douni E, Fiedler IAK, Fonseca JE, Hassler E, Hoegler W, Kague E, Karasik D, Khashayar P, Langdahl BL, Leitch VD, Lopes P, Markozannes G, McGuigan FEA, Medina-Gomez C, Ntzani E, Oei L, Ohlsson C, Szulc P, Tobias JH, Trajanoska K, Tuzun S, Valjevac A, van Rietbergen B, Williams GR, Zekic T, Rivadeneira F, Obermayer-Pietsch Bet al., 2021, Bone phenotyping approaches in human, mice and zebrafish - expert overview of the EU cost action GEMSTONE ("GEnomics of MusculoSkeletal traits TranslatiOnal NEtwork"), Frontiers in Endocrinology, Vol: 12, Pages: 1-32, ISSN: 1664-2392

A synoptic overview of scientific methods applied in bone and associated research fields across species has yet to be published. Experts from the EU Cost Action GEMSTONE (“GEnomics of MusculoSkeletal Traits translational Network”) Working Group 2 present an overview of the routine techniques as well as clinical and research approaches employed to characterize bone phenotypes in humans and selected animal models (mice and zebrafish) of health and disease. The goal is consolidation of knowledge and a map for future research. This expert paper provides a comprehensive overview of state-of-the-art technologies to investigate bone properties in humans and animals – including their strengths and weaknesses. New research methodologies are outlined and future strategies are discussed to combine phenotypic with rapidly developing –omics data in order to advance musculoskeletal research and move towards “personalised medicine”.

Journal article

Youlten SE, Kemp JP, Logan JG, Ghirardello EJ, Sergio CM, Dack MRG, Guilfoyle SE, Leitch VD, Butterfield NC, Komla-Ebri D, Chai RC, Corr AP, Smith JT, Mohanty ST, Morris JA, McDonald MM, Quinn JMW, McGlade AR, Bartonicek N, Jansson M, Hatzikotoulas K, Irving MD, Beleza-Meireles A, Rivadeneira F, Duncan E, Richards JB, Adams DJ, Lelliott CJ, Brink R, Phan TG, Eisman JA, Evans DM, Zeggini E, Baldock PA, Bassett JHD, Williams GR, Croucher PIet al., 2021, Osteocyte transcriptome mapping identifies a molecular landscape controlling skeletal homeostasis and susceptibility to skeletal disease, Nature Communications, Vol: 12, Pages: 1-21, ISSN: 2041-1723

Osteocytes are master regulators of the skeleton. We mapped the transcriptome of osteocytes from different skeletal sites, across age and sexes in mice to reveal genes and molecular programs that control this complex cellular-network. We define an osteocyte transcriptome signature of 1239 genes that distinguishes osteocytes from other cells. 77% have no previously known role in the skeleton and are enriched for genes regulating neuronal network formation, suggesting this programme is important in osteocyte communication. We evaluated 19 skeletal parameters in 733 knockout mouse lines and reveal 26 osteocyte transcriptome signature genes that control bone structure and function. We showed osteocyte transcriptome signature genes are enriched for human orthologs that cause monogenic skeletal disorders (P = 2.4 × 10−22) and are associated with the polygenic diseases osteoporosis (P = 1.8 × 10−13) and osteoarthritis (P = 1.6 × 10−7). Thus, we reveal the molecular landscape that regulates osteocyte network formation and function and establish the importance of osteocytes in human skeletal disease.

Journal article

Dennis EP, Brito FMJDS, Pearson RD, Jackson RM, Logan J, Capulli M, Teti A, Bassett JHD, Williams GR, Kalamajski S, Pirog KAet al., 2021, Asporin is important in bone development but not in cartilage homeostasis, Publisher: WILEY, Pages: A8-A9, ISSN: 0959-9673

Conference paper

Formosa MM, Bergen DJM, Gregson CL, Maurizi A, Kämpe A, Garcia-Giralt N, Zhou W, Grinberg D, Ovejero Crespo D, Zillikens MC, Williams GR, Bassett JHD, Brandi ML, Sangiorgi L, Balcells S, Högler W, Van Hul W, Mäkitie Oet al., 2021, A Roadmap to gene discoveries and novel therapies in monogenic low and high bone mass disorders, Frontiers in Endocrinology, Vol: 12, Pages: 1-24, ISSN: 1664-2392

Genetic disorders of the skeleton encompass a diverse group of bone diseases differing in clinical characteristics, severity, incidence and molecular etiology. Of particular interest are the monogenic rare bone mass disorders, with the underlying genetic defect contributing to either low or high bone mass phenotype. Extensive, deep phenotyping coupled with high-throughput, cost-effective genotyping is crucial in the characterization and diagnosis of affected individuals. Massive parallel sequencing efforts have been instrumental in the discovery of novel causal genes that merit functional validation using in vitro and ex vivo cell-based techniques, and in vivo models, mainly mice and zebrafish. These translational models also serve as an excellent platform for therapeutic discovery, bridging the gap between basic science research and the clinic. Altogether, genetic studies of monogenic rare bone mass disorders have broadened our knowledge on molecular signaling pathways coordinating bone development and metabolism, disease inheritance patterns, development of new and improved bone biomarkers, and identification of novel drug targets. In this comprehensive review we describe approaches to further enhance the innovative processes taking discoveries from clinic to bench, and then back to clinic in rare bone mass disorders. We highlight the importance of cross laboratory collaboration to perform functional validation in multiple model systems after identification of a novel disease gene. We describe the monogenic forms of rare low and high rare bone mass disorders known to date, provide a roadmap to unravel the genetic determinants of monogenic rare bone mass disorders using proper phenotyping and genotyping methods, and describe different genetic validation approaches paving the way for future treatments.

Journal article

Makitie RE, Henning P, Jiu Y, Kampe A, Kogan K, Costantini A, Valimaki V-V, Medina-Gomez C, Pekkinen M, Salusky IB, Schalin-Jantti C, Haanpaa MK, Rivadeneira F, Bassett JHD, Williams GR, Lerner UH, Pereira RC, Lappalainen P, Makitie Oet al., 2021, An ARHGAP25 variant links aberrant Rac1 function to early-onset skeletal fragility, JBMR Plus, Vol: 5, ISSN: 2473-4039

Ras homologous guanosine triphosphatases (RhoGTPases) control several cellular functions, including cytoskeletal actin remodeling and cell migration. Their activities are downregulated by GTPase-activating proteins (GAPs). Although RhoGTPases are implicated in bone remodeling and osteoclast and osteoblast function, their significance in human bone health and disease remains elusive. Here, we report defective RhoGTPase regulation as a cause of severe, early-onset, autosomal-dominant skeletal fragility in a three-generation Finnish family. Affected individuals (n = 13) presented with multiple low-energy peripheral and vertebral fractures despite normal bone mineral density (BMD). Bone histomorphometry suggested reduced bone volume, low surface area covered by osteoblasts and osteoclasts, and low bone turnover. Exome sequencing identified a novel heterozygous missense variant c.652G>A (p.G218R) in ARHGAP25, encoding a GAP for Rho-family GTPase Rac1. Variants in the ARHGAP25 5′ untranslated region (UTR) also associated with BMD and fracture risk in the general population, across multiple genomewide association study (GWAS) meta-analyses (lead variant rs10048745). ARHGAP25 messenger RNA (mRNA) was expressed in macrophage colony-stimulating factor (M-CSF)–stimulated human monocytes and mouse osteoblasts, indicating a possible role for ARHGAP25 in osteoclast and osteoblast differentiation and activity. Studies on subject-derived osteoclasts from peripheral blood mononuclear cells did not reveal robust defects in mature osteoclast formation or resorptive activity. However, analysis of osteosarcoma cells overexpressing the ARHGAP25 G218R-mutant, combined with structural modeling, confirmed that the mutant protein had decreased GAP-activity against Rac1, resulting in elevated Rac1 activity, increased cell spreading, and membrane ruffling. Our findings indicate that mutated ARHGAP25 causes aberrant Rac1 function and consequently abnormal bone metabolism, highlig

Journal article

Butterfield NC, Curry KF, Steinberg J, Dewhurst H, Komla-Ebri D, Mannan NS, Adoum A-T, Leitch VD, Logan JG, Waung JA, Ghirardello E, Southam L, Youlten SE, Wilkinson JM, McAninch EA, Vancollie VE, Kussy F, White JK, Lelliott CJ, Adams DJ, Jacques R, Bianco AC, Boyde A, Zeggini E, Croucher PI, Williams GR, Bassett JHDet al., 2021, Publisher Correction: Accelerating functional gene discovery in osteoarthritis, Nature Communications, Vol: 12, ISSN: 2041-1723

Journal article

McDonald MM, Khoo WH, Ng PY, Xiao Y, Zamerli J, Thatcher P, Kyaw W, Pathmanandavel K, Grootveld AK, Moran I, Butt D, Nguyen A, Corr A, Warren S, Biro M, Butterfield NC, Guilfoyle SE, Komla-Ebri D, Dack MRG, Dewhurst HF, Logan JG, Li Y, Mohanty ST, Byrne N, Terry RL, Simic MK, Chai R, Quinn JMW, Youlten SE, Pettitt JA, Abi-Hanna D, Jain R, Weninger W, Lundberg M, Sun S, Ebetino FH, Timpson P, Lee WM, Baldock PA, Rogers MJ, Brink R, Williams GR, Bassett JHD, Kemp JP, Pavlos NJ, Croucher PI, Phan TGet al., 2021, Osteoclasts recycle via osteomorphs during RANKL-stimulated bone resorption., Cell, Vol: 184

Journal article

McDonald MM, Khoo WH, Ng PY, Xiao Y, Zamerli J, Thatcher P, Kyaw W, Pathmanandavel K, Grootveld AK, Moran I, Butt D, Nguyen A, Corr A, Warren S, Biro M, Butterfield NC, Guilfoyle SE, Komla-Ebri D, Dack MRG, Dewhurst HF, Logan JG, Li Y, Mohanty ST, Byrne N, Terry RL, Simic MK, Chai R, Quinn JMW, Youlten SE, Pettitt JA, Abi-Hanna D, Jain R, Weninger W, Lundberg M, Sun S, Ebetino FH, Timpson P, Lee WM, Baldock PA, Rogers MJ, Brink R, Williams GR, Bassett JHD, Kemp JP, Pavlos NJ, Croucher P, Phan TGet al., 2021, Osteoclasts recycle via osteomorphs during RANKL-stimulated bone resorption, Cell, Vol: 184, Pages: 1330-1347.e13, ISSN: 0092-8674

Osteoclasts are large multinucleated bone-resorbing cells formed by the fusion of monocyte/macrophage-derived precursors that are thought to undergo apoptosis once resorption is complete. Here, by intravital imaging, we reveal that RANKL-stimulated osteoclasts have an alternative cell fate in which they fission into daughter cells called osteomorphs. Inhibiting RANKL blocked this cellular recycling and resulted in osteomorph accumulation. Single-cell RNA sequencing showed that osteomorphs are transcriptionally distinct from osteoclasts and macrophages and express a number of non-canonical osteoclast genes that are associated with structural and functional bone phenotypes when deleted in mice. Furthermore, genetic variation in human orthologs of osteomorph genes causes monogenic skeletal disorders and associates with bone mineral density, a polygenetic skeletal trait. Thus, osteoclasts recycle via osteomorphs, a cell type involved in the regulation of bone resorption that may be targeted for the treatment of skeletal diseases.

Journal article

Steinberg J, Southam L, Roumeliotis T, Clark MJ, Jayasuriya RL, Swift D, Shah KM, Butterfield NC, Brooks RA, McCaskie AW, Bassett JHD, Williams GR, Choudhary JS, Wilkinson JM, Zeggini Eet al., 2021, A molecular quantitative trait locus map for osteoarthritis, Nature Communications, Vol: 12, Pages: 1-11, ISSN: 2041-1723

Osteoarthritis causes pain and functional disability for over 500 million people worldwide. To develop disease-stratifying tools and modifying therapies, we need a better understanding of the molecular basis of the disease in relevant tissue and cell types. Here, we study primary cartilage and synovium from 115 patients with osteoarthritis to construct a deep molecular signature map of the disease. By integrating genetics with transcriptomics and proteomics, we discover molecular trait loci in each tissue type and omics level, identify likely effector genes for osteoarthritis-associated genetic signals and highlight high-value targets for drug development and repurposing. These findings provide insights into disease aetiopathology, and offer translational opportunities in response to the global clinical challenge of osteoarthritis.

Journal article

Tobias JH, Duncan EL, Kague E, Hammond CL, Gregson CL, Bassett D, Williams GR, Min JL, Gaunt TR, Karasik D, Ohlsson C, Rivadeneira F, Edwards JR, Hannan FM, Kemp JP, Gilbert SJ, Alonso N, Hassan N, Compston JE, Ralston SHet al., 2021, Opportunities and challenges in functional genomics research in osteoporosis: report from a workshop held by the causes working group of the osteoporosis and bone research academy of the Royal Osteoporosis Society on October 5th 2020, Frontiers in Endocrinology, Vol: 11, Pages: 1-11, ISSN: 1664-2392

The discovery that sclerostin is the defective protein underlying the rare heritable bone mass disorder, sclerosteosis, ultimately led to development of anti-sclerostin antibodies as a new treatment for osteoporosis. In the era of large scale GWAS, many additional genetic signals associated with bone mass and related traits have since been reported. However, how best to interrogate these signals in order to identify the underlying gene responsible for these genetic associations, a prerequisite for identifying drug targets for further treatments, remains a challenge. The resources available for supporting functional genomics research continues to expand, exemplified by “multi-omics” database resources, with improved availability of datasets derived from bone tissues. These databases provide information about potential molecular mediators such as mRNA expression, protein expression, and DNA methylation levels, which can be interrogated to map genetic signals to specific genes based on identification of causal pathways between the genetic signal and the phenotype being studied. Functional evaluation of potential causative genes has been facilitated by characterization of the “osteocyte signature”, by broad phenotyping of knockout mice with deletions of over 7,000 genes, in which more detailed skeletal phenotyping is currently being undertaken, and by development of zebrafish as a highly efficient additional in vivo model for functional studies of the skeleton. Looking to the future, this expanding repertoire of tools offers the hope of accurately defining the major genetic signals which contribute to osteoporosis. This may in turn lead to the identification of additional therapeutic targets, and ultimately new treatments for osteoporosis.

Journal article

Bassett J, Williams GR, 2021, Accelerating functional gene discovery in osteoarthritis, Nature Communications, Vol: 12, Pages: 1-18, ISSN: 2041-1723

Osteoarthritis causes debilitating pain and disability, resulting in a considerablesocioeconomic burden, yet no drugs are available that prevent disease onset or progression.Here, we develop, validate and use rapid-throughput imaging techniques to identify abnormaljoint phenotypes in randomly selected mutant mice generated by the International KnockoutMouse Consortium. We identify 14 genes with functional involvement in osteoarthritispathogenesis, including the homeobox gene Pitx1, and functionally characterize 6 candidatehuman osteoarthritis genes in mouse models. We demonstrate sensitivity of the methods byidentifying age-related degenerative joint damage in wild-type mice. Finally, we phenotypepreviously generated mutant mice with an osteoarthritis-associated polymorphism in the Dio2gene by CRISPR/Cas9 genome editing and demonstrate a protective role in disease onsetwith public health implications. This expanding resource of mutant mice will acceleratefunctional gene discovery in osteoarthritis and offer drug discovery opportunities for thiscommon, incapacitating chronic disease.

Journal article

Freudenthal B, Watts L, Bassett JHD, Williams Get al., 2021, Thyroid hormone, thyroid medication, and the skeleton, Marcus and Feldman's Osteoporosis (Fifth Edition)

Thyroid hormone is an essential systemic regulator of development and metabolism and has important effects on bone that are mediated principally by thyroid hormone receptor α. In children, hypothyroidism causes growth retardation and delayed bone age, whereas hyperthyroidism accelerates linear growth and advances skeletal maturation. In adults, hyperthyroidism causes high bone turnover osteoporosis and an increased risk of fracture. Overt thyrotoxicosis, subclinical hyperthyroidism, and overtreatment of hypothyroid patients with thyroxine can all result in reduced bone mineral density and an increased susceptibility to fracture. Thyroid hormones are thus essential for normal skeletal development and the normal maintenance of adult bone. When treating patients with thyroid disorders, it is important to consider the potential for detrimental consequences to the skeleton.

Book chapter

Swan AL, Schuett C, Rozman J, Moreno MDMM, Brandmaier S, Simon M, Leuchtenberger S, Griffiths M, Brommage R, Keskivali-Bond P, Grallert H, Werner T, Teperino R, Becker L, Miller G, Moshiri A, Seavitt JR, Cissell DD, Meehan TF, Acar EF, Lelliott CJ, Flenniken AM, Champy M-F, Sorg T, Ayadi A, Braun RE, Cater H, Dickinson ME, Flicek P, Gallegos J, Ghirardello EJ, Heaney JD, Jacquot S, Lally C, Logan JG, Teboul L, Mason J, Spielmann N, McKerlie C, Murray SA, Nutter LMJ, Odfalk KF, Parkinson H, Prochazka J, Reynolds CL, Selloum M, Spoutil F, Svenson KL, Vales TS, Wells SE, White JK, Sedlacek R, Wurst W, Lloyd KKC, Croucher P, Fuchs H, Williams GR, Bassett D, Gailus-Durner V, Herault Y, Mallon A-M, Brown SDM, Mayer-Kuckuk P, de Angelis MHet al., 2020, Mouse mutant phenotyping at scale reveals novel genes controlling bone mineral density, PLoS Genetics, Vol: 16, Pages: 1-27, ISSN: 1553-7390

The genetic landscape of diseases associated with changes in bone mineral density (BMD), such as osteoporosis, is only partially understood. Here, we explored data from 3,823 mutant mouse strains for BMD, a measure that is frequently altered in a range of bone pathologies, including osteoporosis. A total of 200 genes were found to significantly affect BMD. This pool of BMD genes comprised 141 genes with previously unknown functions in bone biology and was complementary to pools derived from recent human studies. Nineteen of the 141 genes also caused skeletal abnormalities. Examination of the BMD genes in osteoclasts and osteoblasts underscored BMD pathways, including vesicle transport, in these cells and together with in silico bone turnover studies resulted in the prioritization of candidate genes for further investigation. Overall, the results add novel pathophysiological and molecular insight into bone health and disease.

Journal article

Manousaki D, Forgetta V, Keller-Baruch J, Zhao K, Greenwood CMT, Mooser V, Bassett JHD, Leslie WD, Richards JBet al., 2020, A Polygenic Risk Score as a Risk Factor for Medication-Associated Fractures, JOURNAL OF BONE AND MINERAL RESEARCH, Vol: 35, Pages: 1935-1941, ISSN: 0884-0431

Journal article

Pereira M, Ko J-H, Logan J, Protheroe H, Kim K-B, Tan ALM, Croucher PI, Park K-S, Rotival M, Petretto E, Bassett JD, Williams GR, Behmoaras Jet al., 2020, A trans-eQTL network regulates osteoclast multinucleation and bone mass, eLife, Vol: 9, ISSN: 2050-084X

Functional characterisation of cell-type-specific regulatory networks is key to establish a causal link between genetic variation and phenotype. The osteoclast offers a unique model for interrogating the contribution of co-regulated genes to in vivo phenotype as its multinucleation and resorption activities determine quantifiable skeletal traits. Here we took advantage of a trans-regulated gene network (MMnet, macrophage multinucleation network) which we found to be significantly enriched for GWAS variants associated with bone-related phenotypes. We found that the network hub gene Bcat1 and seven other co-regulated MMnet genes out of 13, regulate bone function. Specifically, global (Pik3cb-/-, Atp8b2+/-, Igsf8-/-, Eml1-/-, Appl2-/-, Deptor-/-) and myeloid-specific Slc40a1 knockout mice displayed abnormal bone phenotypes. We report opposing effects of MMnet genes on bone mass in mice and osteoclast multinucleation/resorption in humans with strong correlation between the two. These results identify MMnet as a functionally conserved network that regulates osteoclast multinucleation and bone mass.

Journal article

Joustra SD, Roelfsema F, van Trotsenburg ASP, Schneider HJ, Kosilek RP, Kroon HM, Logan JG, Butterfield NC, Zhou X, Toufaily C, Bak B, Turgeon M-O, Brule E, Steyn FJ, Gurnell M, Koulouri O, Le Tissier P, Fontanaud P, Bassett JHD, Williams GR, Oostdijk W, Wit JM, Pereira AM, Biermasz NR, Bernard DJ, Schoenmakers Net al., 2020, Response to Letter to the Editor: "IGSF1 Deficiency Results in Human and Murine Somatotrope Neurosecretory Hyperfunction", JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM, Vol: 105, Pages: E2315-E2316, ISSN: 0021-972X

Journal article

Freudenthal B, Makitie R, Logan J, Sparkes P, Komla Ebri D, Pollard A, Williams G, Bassett Jet al., 2020, A mouse model of juvenile onset X-linked osteoporosis, Bone Research Society BRS Online Rare Bone Disease 2020

Conference paper

Leitch VD, Bassett JHD, Williams GR, 2020, Role of thyroid hormones in craniofacial development, NATURE REVIEWS ENDOCRINOLOGY, Vol: 16, Pages: 147-164, ISSN: 1759-5029

Journal article

Joustra SD, Roelfsema F, van Trotsenburg ASP, Schneider HJ, Kosilek RP, Kroon HM, Logan JG, Butterfield NC, Zhou X, Toufaily C, Bak B, Turgeon M-O, Brule E, Steyn FJ, Gurnell M, Koulouri O, Le Tissier P, Fontanaud P, Bassett JHD, Williams GR, Oostdijk W, Wit JM, Pereira AM, Biermasz NR, Bernard DJ, Schoenmakers Net al., 2020, IGSF1 Deficiency Results in Human and Murine Somatotrope Neurosecretory Hyperfunction, JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM, Vol: 105, Pages: E70-E84, ISSN: 0021-972X

Journal article

Pereira M, Ko J-H, Logan J, Protheroe H, Kim K-B, Tan ALM, Park K-S, Rotival M, Petretto E, Bassett JHD, Williams GR, Behmoaras Jet al., 2020, A trans-eQTL network regulates osteoclast multinucleation and bone mass

<jats:title>Abstract</jats:title><jats:p>Functional characterisation of cell-type specific regulatory networks is key to establish a causal link between genetic variation and phenotype. The osteoclast offers a unique model for interrogating the contribution of co-regulated genes to <jats:italic>in vivo</jats:italic> phenotype as its multinucleation and resorption activities determine quantifiable skeletal traits. Here we took advantage of a <jats:italic>trans</jats:italic>-regulated gene network (MMnet, macrophage multinucleation network) which we found to be significantly enriched for GWAS variants associated with bone-related phenotypes. We found that the network hub gene <jats:italic>Bcat1</jats:italic> and seven other co-regulated MMnet genes out of 13, regulate bone function. Specifically, global (<jats:italic>Pik3cb</jats:italic><jats:sup>−/−</jats:sup>, <jats:italic>Atp8b2</jats:italic><jats:sup>+/−</jats:sup>, <jats:italic>Igsf8</jats:italic><jats:sup>−/−</jats:sup>, <jats:italic>Eml1</jats:italic><jats:sup>−/−</jats:sup>, <jats:italic>Appl2</jats:italic><jats:sup>−/−</jats:sup>, <jats:italic>Deptor</jats:italic><jats:sup>−/−</jats:sup>) and myeloid-specific <jats:italic>Slc40a1<jats:sup>ΔLysMCre</jats:sup></jats:italic> knockout mice displayed abnormal bone phenotypes. We report antagonizing effects of MMnet genes on bone mass in mice and osteoclast multinucleation/resorption in humans with strong correlation between the two. These results identify MMnet as a functionally conserved network that regulates osteoclast fusion and bone mass.</jats:p><jats:sec><jats:title>Impact statement</jats:title><jats:p>We took advantage of the osteoclast w

Working paper

Freudenthal B, Watts L, Williams G, Bassett Jet al., 2020, The skeletal system in thyrotoxicosis, Werner & Ingbar's The Thyroid

Book chapter

Watts L, Freudenthal B, Bassett J, Williams Get al., 2020, The skeletal system in hypothyroidism, Werner & Ingbar's The Thyroid

Book chapter

Laurent L, Zhou S, Forgetta V, Richards JB, Kemp JP, Evans DM, Pramatarova A, Croucher PI, Williams GR, Bassett JHD, Goltzman Det al., 2019, Identification and Validation of Targets for Osteoporosis: Evidence from Whole exome Sequencing in 42,263 individuals, CRISPR-Cas9 and murine models, Annual Meeting of the American-Society-for-Bone-and Mineral Research, Publisher: WILEY, Pages: 28-28, ISSN: 0884-0431

Conference paper

Leitch VD, Brassill MJ, Rahman S, Butterfield NC, Ma P, Logan JG, Boyde A, Evans H, Croucher PI, Batterham RL, Williams GR, Bassett JHDet al., 2019, PYY is a negative regulator of bone mass and strength, Bone, Vol: 127, Pages: 427-435, ISSN: 8756-3282

ObjectiveBone loss in anorexia nervosa and following bariatric surgery is associated with an elevated circulating concentration of the gastrointestinal, anorexigenic hormone, peptide YY (PYY). Selective deletion of the PYY receptor Y1R in osteoblasts or Y2R in the hypothalamus results in high bone mass, but deletion of PYY in mice has resulted in conflicting skeletal phenotypes leading to uncertainty regarding its role in the regulation of bone mass. As PYY analogs are under development for treatment of obesity, we aimed to clarify the relationship between PYY and bone mass.MethodsThe skeletal phenotype of Pyy knockout (KO) mice was investigated during growth (postnatal day P14) and adulthood (P70 and P186) using X-ray microradiography, micro-CT, back-scattered electron scanning electron microscopy (BSE-SEM), histomorphometry and biomechanical testing.ResultsBones from juvenile and Pyy KO mice were longer (P < 0.001), with decreased bone mineral content (P < 0.001). Whereas, bones from adult Pyy KO mice had increased bone mineral content (P < 0.05) with increased mineralisation of both cortical (P < 0.001) and trabecular (P < 0.001) compartments. Long bones from adult Pyy KO mice were stronger (maximum load P < 0.001), with increased stiffness (P < 0.01) and toughness (P < 0.05) compared to wild-type (WT) control mice despite increased cortical vascularity and porosity (P < 0.001). The increased bone mass and strength in Pyy KO mice resulted from increases in trabecular (P < 0.01) and cortical bone formation (P < 0.05).ConclusionsThese findings demonstrate that PYY acts as a negative regulator of osteoblastic bone formation, implicating increased PYY levels in the pathogenesis of bone loss during anorexia or following bariatric surgery.

Journal article

This data is extracted from the Web of Science and reproduced under a licence from Thomson Reuters. You may not copy or re-distribute this data in whole or in part without the written consent of the Science business of Thomson Reuters.

Request URL: http://wlsprd.imperial.ac.uk:80/respub/WEB-INF/jsp/search-html.jsp Request URI: /respub/WEB-INF/jsp/search-html.jsp Query String: respub-action=search.html&id=00344568&limit=30&person=true