Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Brett M. Hosking is active.

Publication


Featured researches published by Brett M. Hosking.


Nature | 2008

Sox18 induces development of the lymphatic vasculature in mice

Mathias Francois; Andrea Caprini; Brett M. Hosking; Fabrizio Orsenigo; Dagmar Wilhelm; Catherine M. Browne; Karri Paavonen; Tara Karnezis; Ramin Shayan; Meredith Downes; Tara Davidson; D. Tutt; Kathryn S. E. Cheah; Steven A. Stacker; George E. O. Muscat; Marc G. Achen; Elisabetta Dejana; Peter Koopman

The lymphatic system plays a key role in tissue fluid regulation and tumour metastasis, and lymphatic defects underlie many pathological states including lymphoedema, lymphangiectasia, lymphangioma and lymphatic dysplasia. However, the origins of the lymphatic system in the embryo, and the mechanisms that direct growth of the network of lymphatic vessels, remain unclear. Lymphatic vessels are thought to arise from endothelial precursor cells budding from the cardinal vein under the influence of the lymphatic hallmark gene Prox1 (prospero homeobox 1; ref. 4). Defects in the transcription factor gene SOX18 (SRY (sex determining region Y) box 18) cause lymphatic dysfunction in the human syndrome hypotrichosis-lymphoedema-telangiectasia, suggesting that Sox18 may also play a role in lymphatic development or function. Here we use molecular, cellular and genetic assays in mice to show that Sox18 acts as a molecular switch to induce differentiation of lymphatic endothelial cells. Sox18 is expressed in a subset of cardinal vein cells that later co-express Prox1 and migrate to form lymphatic vessels. Sox18 directly activates Prox1 transcription by binding to its proximal promoter. Overexpression of Sox18 in blood vascular endothelial cells induces them to express Prox1 and other lymphatic endothelial markers, while Sox18-null embryos show a complete blockade of lymphatic endothelial cell differentiation from the cardinal vein. Our findings demonstrate a critical role for Sox18 in developmental lymphangiogenesis, and suggest new avenues to investigate for therapeutic management of human lymphangiopathies.


Nature Genetics | 2000

Mutations in Sox18 underlie cardiovascular and hair follicle defects in ragged mice.

David J. Pennisi; Jennifer M. Gardner; Doreen Chambers; Brett M. Hosking; Josephine Peters; George E. O. Muscat; Catherine M. Abbott; Peter Koopman

Analysis of classical mouse mutations has been useful in the identification and study of many genes. We previously mapped Sox18, encoding an SRY-related transcription factor, to distal mouse chromosome 2 (ref. 2). This region contains a known mouse mutation, ragged (Ra), that affects the coat and vasculature. Here we have directly evaluated Sox18 as a candidate for Ra. We found that Sox18 is expressed in the developing vascular endothelium and hair follicles in mouse embryos. Furthermore, we found no recombination between Sox18 and Ra in an interspecific backcross segregating for the Ra phenotype. We found point mutations in Sox18 in two different Ra alleles that result in missense translation and premature truncation of the encoded protein. Fusion proteins containing these mutations lack the ability to activate transcription relative to wild-type controls in an in vitro assay. Our observations implicate mutations in Sox18 as the underlying cause of the Ra phenotype, and identify Sox18 as a critical gene for cardiovascular and hair follicle formation.


Development | 2009

Sox7 and Sox17 are strain-specific modifiers of the lymphangiogenic defects caused by Sox18 dysfunction in mice

Brett M. Hosking; Mathias Francois; Dagmar Wilhelm; Fabrizio Orsenigo; Andrea Caprini; Terje Svingen; D. Tutt; Tara Davidson; Catherine M. Browne; Elisabetta Dejana; Peter Koopman

Developmental defects caused by targeted gene inactivation in mice are commonly subject to strain-specific modifiers that modulate the severity of the phenotype. Although several genetic modifier loci have been mapped in mice, the gene(s) residing at these loci are mostly unidentified, and the molecular mechanisms of modifier action remain poorly understood. Mutations in Sox18 cause a variable phenotype in the human congenital syndrome hypotrichosis-lymphedema-telangiectasia, and the phenotype of Sox18-null mice varies from essentially normal to completely devoid of lymphatic vasculature and lethal, depending on the strain of the mice, suggesting a crucial role for strain-specific modifiers in this system. Here we show that two closely related Group F Sox factors, SOX7 and SOX17, are able to functionally substitute for SOX18 in vitro and in vivo. SOX7 and SOX17 are not normally expressed during lymphatic development, excluding a conventional redundancy mechanism. Instead, these genes are activated specifically in the absence of SOX18 function, and only in certain strains. Our studies identify Sox7 and Sox17 as modifiers of the Sox18 mutant phenotype, and reveal their mechanism of action as a novel mode of strain-specific compensatory upregulation.


Gene | 1995

Sequence and expression of Sox-18 encoding a new HMG-box transcription factor

Timothy L. Dunn; Lesley Mynett-Johnson; Edwina Wright; Brett M. Hosking; Peter Koopman; George E. O. Muscat

The newly identified Sox gene family (Sry-like HMG-box gene) is characterized by a conserved DNA sequence encoding a domain of approx. 80 amino acids (aa) which is responsible for sequence-specific DNA binding. The first member isolated, the mammalian Y-linked testis-determining gene, Sry, is necessary and sufficient for male development. We report here the identification of two new members of this family, Sox-17 and 18. We have determined the full cDNA sequence of Sox-18 which encodes a protein of 378 aa. Sox-18 mRNA transcripts were restricted to heart, lung and skeletal muscle in the adult mouse.


Journal of Biological Chemistry | 2004

The VCAM-1 Gene That Encodes the Vascular Cell Adhesion Molecule Is a Target of the Sry-related High Mobility Group Box Gene, Sox18

Brett M. Hosking; S.-C. Mary Wang; Meredith Downes; Peter Koopman; George E. O. Muscat

VCAM-1 (vascular cell adhesion molecule-1) and Sox18 are involved in vascular development. VCAM-1 is an important adhesion molecule that is expressed on endothelial cells and has a critical role in endothelial activation, inflammation, lymphatic pathophysiology, and atherogenesis. The Sry-related high mobility group box factor Sox18 has previously been implicated in endothelial pathologies. Mutations in human and mouse Sox18 leads to hypotrichosis and lymphedema. Furthermore, both Sox18 and VCAM-1 have very similar spatio-temporal patterns of expression, which is suggestive of cross-talk. We use biochemical techniques, cell culture systems, and the ragged opossum (RaOP) mouse model with a naturally occurring mutation in Sox18 to demonstrate that VCAM-1 is an important target of Sox18. Transfection, site-specific mutagenesis, and gel shift analyses demonstrated that Sox18 directly targeted and trans-activated VCAM-1 expression. Importantly, the naturally occurring Sox18 mutant attenuates the expression and activation of VCAM-1 in vitro. Furthermore, in vivo quantitation of VCAM-1 mRNA levels in wild type and RaOP mice demonstrates that RaOP animals show a dramatic and significant reduction in VCAM-1 mRNA expression in lung, skin, and skeletal muscle. Our observation that the VCAM-1 gene is an important target of SOX18 provides the first molecular insights into the vascular abnormalities in the mouse mutant ragged and the human hypotrichosislymphedema-telangiectasia disorder.


Developmental Dynamics | 2009

Ex vivo magnetofection: A novel strategy for the study of gene function in mouse organogenesis

Terje Svingen; Dagmar Wilhelm; Alexander N. Combes; Brett M. Hosking; Vincent R. Harley; Andrew H. Sinclair; Peter Koopman

Gene function during mouse development is often studied through the production and analysis of transgenic and knockout models. However, these techniques are time‐ and resource‐consuming, and require specialized equipment and expertise. We have established a new protocol for functional studies that combines organ culture of explanted fetal tissues with microinjection and magnetically induced transfection (“magnetofection”) of gene expression constructs. As proof‐of‐principle, we magnetofected cDNA constructs into genital ridge tissue as a means of gain‐of‐function analysis, and shRNA constructs for loss‐of‐function analysis. Ectopic expression of Sry induced female‐to‐male sex‐reversal, whereas knockdown of Sox9 expression caused male‐to‐female sex‐reversal, consistent with the known functions of these genes. Furthermore, ectopic expression of Tmem184a, a gene of unknown function, in female genital ridges, resulted in failure of gonocytes to enter meiosis. This technique will likely be applicable to the study of gene function in a broader range of developing organs and tissues. Developmental Dynamics 238:956–964, 2009.


Mammalian Genome | 2000

Structure, mapping, and expression of human SOX18

David J. Pennisi; Kristy M. James; Brett M. Hosking; George E. O. Muscat; Peter Koopman

SOX genes are found throughout the animal kingdom and encode a highly conserved family of transcription factors involved in a wide range of developmental processes (for review, see Wegner, 1999). SOX proteins bind DNA in a sequence-dependent manner via the HMG box domain, and many have been shown to have separable domains associated with transcriptional regulation. Several SOX genes have been implicated in inherited human disorders: SRY in male-to-female sex reversal and gonadal dysgenesis (Berta et al. 1990; Ja ger et al. 1990), SOX9 in the skeletal dys-morphogenesis syndrome campomelic dysplasia (Foster and Graves 1994; Wagner et al. 1994), and SOX10 in the neurochristopathy Waardenburg Shah syndrome-4 (Pingault et al. 1998). Furthermore, targeted disruption in mice has demonstrated vital roles for Sox4 in cardiac development (Schilham et al. 1996), Sox9 in chondrogenesis (Bi et al. 1999), and Sox1 in lens development (Nishiguchi et al. 1998).


Genes & Development | 2000

The steroid receptor coactivator, GRIP-1, is necessary for MEF-2C-dependent gene expression and skeletal muscle differentiation

Shen Liang Chen; Dennis H. Dowhan; Brett M. Hosking; George E. O. Muscat


Nucleic Acids Research | 1995

Trans-activation and DNA-binding properties of the transcription factor, Sox-18

Brett M. Hosking; George E. O. Muscat; Peter Koopman; Dennis H. Dowhan; Timothy L. Dunn


Nucleic Acids Research | 2000

Cloning and characterisation of the Sry-related transcription factor gene Sox8

Goslik Schepers; Monica Bullejos; Brett M. Hosking; Peter Koopman

Collaboration


Dive into the Brett M. Hosking's collaboration.

Top Co-Authors

Avatar

Peter Koopman

University of Queensland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

S-C.Mary Wang

University of Queensland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shen Liang Chen

National Central University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

D. Tutt

University of Queensland

View shared research outputs
Researchain Logo
Decentralizing Knowledge