Biology:Brachyury

From HandWiki
A representation of the 3D structure of the protein myoglobin showing turquoise α-helices.
Generic protein structure example

Brachyury (from Greek βραχύς, "short" and ουρά, "tail") is a protein that, in humans, is encoded by the TBXT (T-box transcription factor T) gene.[1][2] Brachyury functions as a transcription factor within the T-box family of genes.[3] Brachyury homologs have been found in all bilaterian animals that have been screened, as well as the freshwater cnidarian Hydra.[3]

History

The brachyury mutation was first described in mice by Nadezhda Alexandrovna Dobrovolskaya-Zavadskaya in 1927 as a mutation that affected tail length and sacral vertebrae in heterozygous animals. In homozygous animals the brachyury mutation is lethal at around embryonic day 10 due to defects in mesoderm formation, notochord differentiation and the absence of structures posterior to the forelimb bud (Dobrovolskaïa-Zavadskaïa, 1927). The name brachyury comes from the Greek brakhus meaning short and oura meaning tail.

In 2018 HGNC updated the human gene name from T to TBXT, presumably to overcome difficulties associated with searching for a single letter gene symbol. It is assumed that the mouse nomenclature will also be updated in due course.

The mouse T gene was cloned by Bernhard Herrmann and colleagues[4] and proved to encode a 436 amino acid embryonic nuclear transcription factor. T binds to a specific DNA element, a near palindromic sequence TCACACCT through a region in its N-terminus, called the T-box. T is the founding member of the T-box family which in mammals currently consists of 18 T-box genes.

The crystal structure of the human brachyury protein was solved in 2017 by Opher Gileadi and colleagues at the Structural Genomics Consortium in Oxford.[5]

Brachyury expression in 7.5dpc CD1 mouse embryos

Role in development

The gene brachyury appears to have a conserved role in defining the midline of a bilaterian organism,[6] and thus the establishment of the anterior-posterior axis; this function is apparent in chordates and molluscs.[7] Its ancestral role, or at least the role it plays in the Cnidaria, appears to be in defining the blastopore.[3] It also defines the mesoderm during gastrulation.[8] Tissue-culture based techniques have demonstrated one of its roles may be in controlling the velocity of cells as they leave the primitive streak.[9][10] It effects transcription of genes required for mesoderm formation and cellular differentiation.[clarification needed]

Brachyury has also been shown to help establish the cervical vertebral blueprint during fetal development. The number of cervical vertebrae is highly conserved among all mammals; however a spontaneous vertebral and spinal dysplasia (VSD) mutation in this gene has been associated with the development of six or fewer cervical vertebrae instead of the usual seven.[11]

Expression

In mice T is expressed in the inner cell mass of the blastocyst stage embryo (but not in the majority of mouse embryonic stem cells) followed by the primitive streak (see image). In later development expression is localised to the node and notochord.

In Xenopus laevis Xbra (the Xenopus T homologue, also recently renamed t) is expressed in the mesodermal marginal zone of the pre-gastrula embryo followed by localisation to the blastopore and notochord at the mid-gastrula stage.

Orthologs

The Danio rerio ortholog is known as ntl (no tail)

Role in disease

Cancer

Brachyury is implicated in the initiation and/or progression of a number of tumor types including chordoma, germ cell tumors, hemangioblastoma, GIST, lung cancer, small cell carcinoma of the lung, breast cancer, colon cancer, hepatocellular carcinoma, prostate cancer, and oral squamous carcinoma.[12]

In breast cancer brachyury expression is associated with recurrence, metastasis and reduced survival.[13][14][15][16] It is also associated with resistance to tamoxifen[17] and to cytotoxic chemotherapy.[13]

In lung cancer brachyury expression is associated with recurrence and decreased survival.[18][19][20][21] It is also associated with resistance to cytotoxic chemotherapy,[22] radiation,[23] and EGFR kinase inhibitors.[18]

In prostate cancer brachyury expression is associated with Gleason score, perineural, invasion and capsular invasion.[24]

In addition to its role in common cancers, brachyury has been identified as a definitive diagnostic marker, key driver and therapeutic target for chordoma, a rare malignant tumor that arises from remnant notochordal cells lodged in the vertebrae. The evidence regarding brachyury's role in chordoma includes:

  • Brachyury is highly expressed in all chordomas except for the dedifferentiated subtype, which accounts for less than 5% of cases[25]
  • Germ line duplication of the brachyury gene is responsible for familial chordoma.[26]
  • A germline SNP in brachyury is present in 97% of chordoma patients.[27]
  • Somatic amplifications of brachyury are seen in a subset of sporadic chordomas either by aneuploidy or focal duplication.[28]
  • Brachyury is the most selectively essential gene in chordoma relative to other cancer types.[29]
  • Brachyury is associated with a large superenhancer in chordoma tumors and cell lines, and is the most highly expressed superenhancer-associated transcription factor.[29]

Brachyury is an important factor in promoting the epithelial–mesenchymal transition (EMT). Cells that over-express brachyury have down-regulated expression of the adhesion molecule E-cadherin, which allows them to undergo EMT. This process is at least partially mediated by the transcription factors AKT[30] and Snail.[31]

Overexpression of brachyury has been linked to hepatocellular carcinoma (HCC, also called malignant hepatoma), a common type of liver cancer. While brachyury is promoting EMT, it can also induce metastasis of HCC cells. Brachyury expression is a prognostic biomarker for HCC, and the gene may be a target for cancer treatments in the future.[30]

Other diseases

Overexpression of brachyury may play a part in EMT associated with benign disease such as renal fibrosis.[31]

Role as a therapeutic target

Because brachyury is expressed in tumors but not in normal adult tissues it has been proposed as a potential drug target with applicability across tumor types. In particular, brachyury-specific peptides are presented on HLA receptors of cells in which it is expressed, representing a tumor specific antigen. Various therapeutic vaccines have been developed which are intended to stimulate an immune response to brachyury expressing cells.[12]

See also


References

  1. "Entrez Gene: T". https://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=6862. 
  2. "The human homolog T of the mouse T(Brachyury) gene; gene structure, cDNA sequence, and assignment to chromosome 6q27". Genome Research 6 (3): 226–33. March 1996. doi:10.1101/gr.6.3.226. PMID 8963900. 
  3. 3.0 3.1 3.2 "The ancestral role of Brachyury: expression of NemBra1 in the basal cnidarian Nematostella vectensis (Anthozoa)". Development Genes and Evolution 212 (12): 563–70. January 2003. doi:10.1007/s00427-002-0272-x. PMID 12536320. 
  4. "Cloning of the T gene required in mesoderm formation in the mouse". Nature 343 (6259): 617–22. February 1990. doi:10.1038/343617a0. PMID 2154694. Bibcode1990Natur.343..617H. 
  5. "Crystal structure of human Brachyury (T) in complex with DNA". To be Published. 2017. doi:10.2210/pdb6f58/pdb. https://www.rcsb.org/structure/6F58. 
  6. "Expression of a SoxB and a Wnt2/13 gene during the development of the mollusc Patella vulgata". Development Genes and Evolution 214 (5): 250–6. May 2004. doi:10.1007/s00427-004-0399-z. PMID 15034714. 
  7. "Expression pattern of Brachyury in the mollusc Patella vulgata suggests a conserved role in the establishment of the AP axis in Bilateria". Development 129 (6): 1411–21. March 2002. PMID 11880350. 
  8. "Evolution of Brachyury proteins: identification of a novel regulatory domain conserved within Bilateria". Developmental Biology 260 (2): 352–61. August 2003. doi:10.1016/S0012-1606(03)00244-6. PMID 12921737. 
  9. "An ECM substratum allows mouse mesodermal cells isolated from the primitive streak to exhibit motility similar to that inside the embryo and reveals a deficiency in the T/T mutant cells". Development 100 (4): 587–98. August 1987. PMID 3327671. http://dev.biologists.org/content/100/4/587. 
  10. "Brachyury cooperates with Wnt/β-catenin signalling to elicit primitive-streak-like behaviour in differentiating mouse embryonic stem cells". BMC Biology 12 (1): 63. August 2014. doi:10.1186/s12915-014-0063-7. PMID 25115237. 
  11. "The mammalian cervical vertebrae blueprint depends on the T (brachyury) gene". Genetics 199 (3): 873–83. March 2015. doi:10.1534/genetics.114.169680. PMID 25614605. 
  12. 12.0 12.1 "Development of Cancer Vaccines Targeting Brachyury, a Transcription Factor Associated with Tumor Epithelial-Mesenchymal Transition". Cells Tissues Organs 203 (2): 128–138. 2017. doi:10.1159/000446495. PMID 28214895. 
  13. 13.0 13.1 "Overexpression of the EMT driver brachyury in breast carcinomas: association with poor prognosis". Journal of the National Cancer Institute 106 (5). May 2014. doi:10.1093/jnci/dju054. PMID 24815864. 
  14. "The potential role of Brachyury in inducing epithelial-to-mesenchymal transition (EMT) and HIF-1α expression in breast cancer cells". Biochemical and Biophysical Research Communications 467 (4): 1083–9. November 2015. doi:10.1016/j.bbrc.2015.09.076. PMID 26393908. 
  15. "Brachyury, a vaccine target, is overexpressed in triple-negative breast cancer". Endocrine-Related Cancer 23 (10): 783–796. October 2016. doi:10.1530/ERC-16-0037. PMID 27580659. 
  16. "Prognostic significance of expression of epithelial-mesenchymal transition driver brachyury in breast cancer and its association with subtype and characteristics". Oncology Letters 15 (1): 1037–1045. January 2018. doi:10.3892/ol.2017.7402. PMID 29399164. 
  17. "Brachyury promotes tamoxifen resistance in breast cancer by targeting SIRT1". Biomedicine & Pharmacotherapy 84: 28–33. December 2016. doi:10.1016/j.biopha.2016.09.011. PMID 27621036. 
  18. 18.0 18.1 "Brachyury, a driver of the epithelial–mesenchymal transition, is overexpressed in human lung tumors: an opportunity for novel interventions against lung cancer". Clinical Cancer Research 18 (14): 3868–79. July 2012. doi:10.1158/1078-0432.CCR-11-3211. PMID 22611028. 
  19. "Expression of Brachyury gene is a significant prognostic factor for primary lung carcinoma". Annals of Surgical Oncology 20 Suppl 3: S509-16. December 2013. doi:10.1245/s10434-013-2914-9. PMID 23456319. 
  20. "Nuclear Brachyury Expression Is Consistent in Chordoma, Common in Germ Cell Tumors and Small Cell Carcinomas, and Rare in Other Carcinomas and Sarcomas: An Immunohistochemical Study of 5229 Cases". The American Journal of Surgical Pathology 39 (10): 1305–12. October 2015. doi:10.1097/PAS.0000000000000462. PMID 26099010. 
  21. "Prognostic Significance of Expression of the Epithelial–Mesenchymal Transition-Related Factor Brachyury in Intrathoracic Lymphatic Spread of Non-Small Cell Lung Cancer". Annals of Surgical Oncology 23 (Suppl 5): 1012–1020. December 2016. doi:10.1245/s10434-016-5530-7. PMID 27600618. 
  22. "Impact of Brachyury on epithelial-mesenchymal transitions and chemosensitivity in non-small cell lung cancer". Molecular Medicine Reports 12 (1): 995–1001. July 2015. doi:10.3892/mmr.2015.3348. PMID 25683840. 
  23. "The embryonic transcription factor Brachyury blocks cell cycle progression and mediates tumor resistance to conventional antitumor therapies". Cell Death & Disease 4 (6): e682. June 2013. doi:10.1038/cddis.2013.208. PMID 23788039. 
  24. "T-box transcription factor brachyury is associated with prostate cancer progression and aggressiveness". Clinical Cancer Research 20 (18): 4949–61. September 2014. doi:10.1158/1078-0432.CCR-14-0421. PMID 25009296. 
  25. "Brachyury, a crucial regulator of notochordal development, is a novel biomarker for chordomas". The Journal of Pathology 209 (2): 157–65. June 2006. doi:10.1002/path.1969. PMID 16538613. 
  26. "T (brachyury) gene duplication confers major susceptibility to familial chordoma". Nature Genetics 41 (11): 1176–8. November 2009. doi:10.1038/ng.454. PMID 19801981. 
  27. "A common single-nucleotide variant in T is strongly associated with chordoma". Nature Genetics 44 (11): 1185–7. November 2012. doi:10.1038/ng.2419. PMID 23064415. 
  28. "The driver landscape of sporadic chordoma". Nature Communications 8 (1): 890. October 2017. doi:10.1038/s41467-017-01026-0. PMID 29026114. Bibcode2017NatCo...8..890T. 
  29. 29.0 29.1 "Small-molecule targeting of brachyury transcription factor addiction in chordoma". Nature Medicine 25 (2): 292–300. February 2019. doi:10.1038/s41591-018-0312-3. PMID 30664779. 
  30. 30.0 30.1 "Overexpression of brachyury contributes to tumor metastasis by inducing epithelial–mesenchymal transition in hepatocellular carcinoma". Journal of Experimental & Clinical Cancer Research 33: 105. December 2014. doi:10.1186/s13046-014-0105-6. PMID 25499255. 
  31. 31.0 31.1 "The T-box transcription factor Brachyury promotes renal interstitial fibrosis by repressing E-cadherin expression". Cell Communication and Signaling 12: 76. November 2014. doi:10.1186/s12964-014-0076-4. PMID 25433496. 

Further reading

External links