top of page

Barsi Lab Research

Dr. Julius Barsi’s research takes a systems biology approach to understanding and explaining the mechanisms by which genetic information leads to anatomical structure. By systematically collecting cell specific data on gene expression he is able to catalogue differentially expressed genes and gene regulatory networks throughout purple sea urchin (Strongylocentrotus purpuratus) embryonic development. Once completed, a catalogue such as this may enable the reverse engineering of sea urchin development for the first time.

Dr. Barsi has reached the stage in S. purpuratus development where the Archenteron cells become highly specialised and will soon differentiate into various gut cells. Data has been collected on Archenteron cells as well as a transcriptome of the entire embryo during mid-gastrulation (30 hpf). This data will be analysed during the internship.

to read about the Barsi Lab visit: https://barsi-leidenfrost.org/

Barsi Lab Research
Data Collection

Data Collection

Many steps have gone into the data collection by Dr. Barsi at at the Bermuda Institute for Ocean Sciences (the procedure is described by Barsi et al. (2014)). These steps are roughly laid here:

1    Create genetically engineered artificial chromosomes whose expression defines a              unique cell type (Fox A in the Archenteron), and inject these into sea urchin embryos.

2    Cultivate ~8000 embryos & disaggregate into single cells at the desired developmental

      stage (here 30hpf).

3    Using fluoresence-activated cell sorting to sort the GFP+ (cell specific) from the GFP-        (control) cells. 

4    Following rolling circle amplification and next generation sequencing library                        preparation, conduct Illumina RNA sequencing on both populations of cells and                    calculate relative enrichment. 

5    Normalising data across biological replicates and for gene length. 

The Purple Sea Urchin

The Purple Sea Urchin

Strongylocentrotus purpuratus

Sea urchins are model organisms commonly used for this type of research for various reasons, hence the purple sea urchins gene regulatory network is one of the best understood at the moment. The main advantage that purple sea urchins provide is their ability to integrate foreign DNA, introduced at the single cell stage, into their own DNA.

 

The images below illustrate their embryonic development, highlighting the stage and tissue of interest:

Capture.JPG

Green and Batterman, 2017. Stud. His. Philos. Sci. A. Part C, 61, pp.20-34

Green and Batterman, 2017. Stud. His. Philos. Sci. A. Part C, 61, pp.20-34

Ettensohn, 2020. Mech. Dev., 162, p103599

Capture2.JPG
Signalling Genes

Signalling Genes

The Internship Focus

Signalling genes are genes that specifically have a role in signalling processes by encoding for the production of receptors and signalling ligands. This allows for communication between cells which is crucial throughout embryonic development as it provides each cell with information on its surroundings and location during this process. Development is controlled by gene regulation of transcription factors and signalling genes (Levine and Davidson, 2005), where gene regulation refers to the process of regulating which subset of genes should be expressed in each cell via transcription factors (Davidson, 2010). The transcription factors present in a cell nucleus determines a cells regulatory state and actively regulates gene expression, and the signalling gene products then allow for communication between cells to ensure each cell is in the correct regulatory state (Davidson, 2010; Cui et al., 2014). Gene regulation occurs as a network, where transcription factors lie upstream of all other genes, meaning that the expression of transcription factors cascades into a network of gene activation and suppression (Davidson, 2010). Signalling genes fit into this network by communicating between the gene regulatory networks of different tissues.

 

Signalling Types in Sea Urchin Embryonic Development

Signalling can either occur short-range or long-range, and during early embryonic development the short-range signalling types are utilised. These consist of contact-dependant, paracrine, and autocrine signalling. Paracrine signalling, where two nearby cells that are not in direct contact communicate, is most often used in sea urchin development around 30hpf. Once a signal is received in a cell it typically triggers a cascade of reactions, i.e. a signalling pathway, that ultimately achieves the goal of the signal. The main pathways used throughout embryonic development in sea urchins are: hedgehog, Wnt, TGF-beta, FGF, RTK, and Notch which all use paracrine signalling with the exception of Notch signalling which uses contact-dependent signalling (e.g. Basson, 2012; Egana and Ernst, 2004; Lapraz et al., 2006; Sanz-Ezquerro et al., 2017; Walton et al., 2006). There are multiple pathways that fall under each of these main ones. Wnt-signalling pathways for example include the canonical Wnt/B-catenin pathway, and the non-canonical WNT/planar cell polarity (PCP) and WNT/ Ca 2+ pathways (Sanz-Ezquerro et al., 2017).

Literature

Literature

Adomako‐Ankomah, A. and Ettensohn, C.A., 2014. Growth factors and early mesoderm morphogenesis: insights from the sea urchin embryo. genesis, 52(3), pp.158-172.

 

Adomako-Ankomah, A. and Ettensohn, C.A., 2013. Growth factor-mediated mesodermal cell guidance and skeletogenesis during sea urchin gastrulation. Development, 140(20), pp.4214-4225.                                                                                                                                              

Andermarcher, E., Surani, M.A. and Gherardi, E., 1996. Co‐expression of the HGF/SF and c‐met genes during early mouse embryogenesis precedes reciprocal expression in adjacent tissues during organogenesis. Developmental genetics, 18(3), pp.254-266.

 

Barsi Lab, 2021. Barsi Lab. [online] Available at: <https://barsi-leidenfrost.org/> [Accessed 29 September 2021].

 

Barsi, J.C., Tu, Q., Calestani, C. and Davidson, E.H., 2015. Genome-wide assessment of differential effector gene use in embryogenesis. Development, 142(22), pp.3892-3901.

 

Barsi, J.C., Tu, Q. and Davidson, E.H., 2014. General approach for in vivo recovery of cell type-specific effector gene sets. Genome research, 24(5), pp.860-868.

 

Basson, M.A., 2012. Signaling in cell differentiation and morphogenesis. Cold Spring Harbor perspectives in biology, 4(6), p.a008151.

 

Biotapestry.org, 2021. BioTapestry Echinolab page on Davidson Lab gene regulatory networks [online] Available at: <http://www.biotapestry.org/> [Accessed 29 September 2021].

 

Braun, T. and Woollard, A., 2009. RUNX factors in development: lessons from invertebrate model systems. Blood Cells, Molecules, and Diseases, 43(1), pp.43-48.

 

Butte, A.J., Ye, J., Häring, H.U., Stumvoll, M., White, M.F. and Kohane, I.S., 2000. Determining significant fold differences in gene expression analysis. In Biocomputing 2001 (pp. 6-17).

 

Chauhan, B.K., Disanza, A., Choi, S.Y., Faber, S.C., Lou, M., Beggs, H.E., Scita, G., Zheng, Y. and Lang, R.A., 2009. Cdc42-and IRSp53-dependent contractile filopodia tether presumptive lens and retina to coordinate epithelial invagination. Development, 136(21), pp.3657-3667

 

Chen, Y., McCarthy, D., Ritchie, M., Robinson, M., Smyth, G. and Hall, E., 2020. edgeR: differential analysis of sequence read count data User’s Guide. Accessed: Jul, 8. http://www.bioconductor.org/packages/release/bioc/vignettes/edgeR/inst/doc/edgeRUsersGuide.pdf

 

Croce, J., Duloquin, L., Lhomond, G., McClay, D.R. and Gache, C., 2006. Frizzled5/8 is required in secondary mesenchyme cells to initiate archenteron invagination during sea urchin development. Development, 131(3), pp.547-557.

 

Croce, J.C. and McClay, D.R., 2010. Dynamics of Delta/Notch signaling on endomesoderm segregation in the sea urchin embryo. Development, 137(1), pp.83-91.

 

Croce, J., Range, R., Wu, S.Y., Miranda, E., Lhomond, G., Peng, J.C.F., Lepage, T. and McClay, D.R., 2011. Wnt6 activates endoderm in the sea urchin gene regulatory network. Development, 138(15), pp.3297-3306.

 

Cui, M., Siriwon, N., Li, E., Davidson, E.H. and Peter, I.S., 2014. Specific functions of the Wnt signaling system in gene regulatory networks throughout the early sea urchin embryo. Proceedings of the National Academy of Sciences, 111(47), pp.E5029-E5038.

 

Davidson, E.H., 2010. The regulatory genome: gene regulatory networks in development and evolution. Elsevier.

 

Egana, A.L. and Ernst, S.G., 2004. Sphedgehog is expressed by pigment cell precursors during early gastrulation in Strongylocentrotus purpuratus. Developmental dynamics: an official publication of the American Association of Anatomists, 231(2), pp.370-378.

 

Gaudino, G., Follenzi, A., Naldini, L., Collesi, C., Santoro, M., Gallo, K.A., Godowski, P.J. and Comoglio, P.M., 1994. RON is a heterodimeric tyrosine kinase receptor activated by the HGF homologue MSP. The EMBO journal, 13(15), pp.3524-3532.

 

Govindarajan, V., Ramachandran, R.K., George, J.M., Shakes, D.C. and Tomlinson, C.R., 1995. An ECM-Bound, PDGF-like Growth Factor and a TGF-α-like Growth Factor Are Required for Gastrulation and Spiculogenesis in the Lytechinus Embryo. Developmental biology, 172(2), pp.541-551.            

 

Greenwood, J. and Gautier, J., 2005, April. From oogenesis through gastrulation: developmental regulation of apoptosis. In Seminars in cell & developmental biology (Vol. 16, No. 2, pp. 215-224)

 

Hamada, M. and Kiyomoto, M., 2003. Signals from primary mesenchyme cells regulate endoderm differentiation in the sea urchin embryo. Development, growth & differentiation, 45(4), pp.339-350.

 

Kikuchi, Y., Verkade, H., Reiter, J.F., Kim, C.H., Chitnis, A.B., Kuroiwa, A. and Stainier, D.Y., 2004. Notch signaling can regulate endoderm formation in zebrafish. Developmental dynamics: an official publication of the American Association of Anatomists, 229(4), pp.756-762

                                                                                                              

Kumano, M. and Foltz, K.R., 2003. Inhibition of mitogen activated protein kinase signaling affects gastrulation and spiculogenesis in the sea urchin embryo. Development, growth & differentiation, 45(5‐6), pp.527-542.

 

Latimer, A.J. and Jessen, J.R., 2008. hgf/c‐met expression and functional analysis during zebrafish embryogenesis. Developmental dynamics: an official publication of the American Association of Anatomists, 237(12), pp.3904-3915.

 

Lapraz, F., Röttinger, E., Duboc, V., Range, R., Duloquin, L., Walton, K., Wu, S.Y., Bradham, C., Loza, M.A., Hibino, T. and Wilson, K., 2006. RTK and TGF-β signaling pathways genes in the sea urchin genome. Developmental biology, 300(1), pp.132-152.

 

Legacy.echinobase.org, 2021. Quantitative Developmental Transcriptomes of S. purpuratus. [online] Available at: <http://legacy.echinobase.org/shiny/quantdev/> [Accessed 29 September 2021].

 

Lehar, S.M. and Bevan, M.J., 2006. T cells develop normally in the absence of both Deltex1 and Deltex2. Molecular and cellular biology, 26(20), pp.7358-7371. 

 

Levine, M. and Davidson, E.H., 2005. Gene regulatory networks for development. Proceedings of the National Academy of Sciences, 102(14), pp.4936-4942.

 

Lhomond, G., McClay, D.R., Gache, C. and Croce, J.C., 2012. Frizzled1/2/7 signaling directs β-catenin nuclearisation and initiates endoderm specification in macromeres during sea urchin embryogenesis. Development, 139(4), pp.816-825.

 

Materna, S.C., Swartz, S.Z. and Smith, J., 2013. Notch and Nodal control forkhead factor expression in the specification of multipotent progenitors in sea urchin. Development, 140(8), pp.1796-1806.

 

McCauley, B.S., Akyar, E., Filliger, L. and Hinman, V.F., 2013. Expression of wnt and frizzled genes during early sea star development. Gene Expression Patterns, 13(8), pp.437-444.

 

McCauley, B.S., Weideman, E.P. and Hinman, V.F., 2010. A conserved gene regulatory network subcircuit drives different developmental fates in the vegetal pole of highly divergent echinoderm embryos. Developmental biology, 340(2), pp.200-208.                                        

Muraoka, R.S., Sun, W.Y., Colbert, M.C., Waltz, S.E., Witte, D.P., Degen, J.L. and Degen, S.J.F., 1999. The Ron/STK receptor tyrosine kinase is essential for peri-implantation development in the mouse. The Journal of clinical investigation, 103(9), pp.1277-1285.  

 

Oliveri, P., Tu, Q. and Davidson, E.H., 2008. Global regulatory logic for specification of an embryonic cell lineage. Proceedings of the National Academy of Sciences, 105(16), pp.5955-5962.

 

Organ, S.L. and Tsao, M.S., 2011. An overview of the c-MET signaling pathway. Therapeutic advances in medical oncology, 3(1_suppl), pp.S7-S19.

 

Robertson, A.J., Coluccio, A., Knowlton, P., Dickey-Sims, C. and Coffman, J.A., 2008. Runx expression is mitogenic and mutually linked to Wnt activity in blastula-stage sea urchin embryos. PloS one, 3(11), p.e3770.

 

Robertson, A.J., Dickey, C.E., McCarthy, J.J. and Coffman, J.A., 2002. The expression of SpRunt during sea urchin embryogenesis. Mechanisms of development, 117(1-2), pp.327-330.

 

Sanz-Ezquerro, J.J., Münsterberg, A.E. and Stricker, S., 2017. Signaling pathways in embryonic development. Frontiers in cell and developmental biology, 5, p.76.

 

Scamuffa, N., Calvo, F., Chrétien, M., Seidah, N.G. and Khatib, A.M., 2006. Proprotein convertases: lessons from knockouts. The FASEB Journal, 20(12), pp.1954-1963.)  

 

Sherwood, D.R. and McClay, D.R., 1997. Identification and localization of a sea urchin Notch homologue: insights into vegetal plate regionalization and Notch receptor regulation. Development, 124(17), pp.3363-3374.  

 

Sun, Z. and Ettensohn, C.A., 2014. Signal-dependent regulation of the sea urchin skeletogenic gene regulatory network. Gene Expression Patterns, 16(2), pp.93-103     

 

Turpeinen, H., Oksanen, A., Kivinen, V., Kukkurainen, S., Uusimäki, A., Rämet, M., Parikka, M., Hytönen, V.P., Nykter, M. and Pesu, M., 2013. Proprotein convertase subtilisin/kexin type 7 (PCSK7) is essential for the zebrafish development and bioavailability of transforming growth factor β1a (TGFβ1a). Journal of Biological Chemistry, 288(51), pp.36610-36623.      

   

van Loo, G., Saelens, X., Van Gurp, M., MacFarlane, M., Martin, S.J. and Vandenabeele, P., 2002. The role of mitochondrial factors in apoptosis: a Russian roulette with more than one bullet. Cell Death & Differentiation, 9(10), pp.1031-1042

 

Wagh, P.K., Gray, J.K., Zinser, G.M., Vasiliauskas, J., James, L., Monga, S.P. and Waltz, S.E., 2011. β-Catenin is required for Ron receptor-induced mammary tumorigenesis. Oncogene, 30(34), pp.3694-3704)   

 

Wagh, P.K., Peace, B.E. and Waltz, S.E., 2008. Met‐related receptor tyrosine kinase Ron in tumor growth and metastasis. Advances in cancer research, 100, pp.1-33.

 

Walton, K.D., Croce, J.C., Glenn, T.D., Wu, S.Y. and McClay, D.R., 2006. Genomics and expression profiles of the Hedgehog and Notch signaling pathways in sea urchin development. Developmental biology, 300(1), pp.153-164.       

 

Wang, Z., Dai, Z., Pan, Y., Wu, S., Li, Z. and Zuo, C., 2017. E3 ubiquitin ligase DTX4 is required for adipogenic differentiation in 3T3-L1 preadipocytes cell line. Biochemical and biophysical research communications, 492(3), pp.419-424.

 

Wang, H.C., Perry, S.S. and Sun, X.H., 2009. Id1 attenuates Notch signaling and impairs T-cell commitment by elevating Deltex1 expression. Molecular and cellular biology, 29(17), pp.4640-4652.

 

Yaguchi, S., 2019. Temnopleurus as an emerging echinoderm model. Methods in cell biology, 150, pp.71-79.

bottom of page