ZFIN ID: ZDB-LAB-100302-2
Hogan Lab
PI/Director: Hogan, Ben M.
Contact Person: Hogan, Ben M.
Email: Ben.Hogan@petermac.org
URL: https://biomedicalsciences.unimelb.edu.au/sbs-research-groups/anatomy-and-physiology-research/stem-cell-and-developmental-biology/hogan-laboratory-vascular-developmental-genetics-and-cell-biology
Address: Organogenesis and Cancer Program Peter MacCallum Cancer Centre 305 Grattan St, Melbourne VIC 3000
Country: Australia
Phone: (+61) 7 3346 2100
Fax: (+61) 7 3346 2101
Line Designation: uom


GENOMIC FEATURES ORIGINATING FROM THIS LAB
Show all 33 genomic features


STATEMENT OF RESEARCH INTERESTS
Understanding lymphatic vessel development using model organism genetics and genomics

Lymphatic vessels form from pre-existing vessels via lymphangiogenesis in both development and in pathological settings. Lymphatics function to drain tissue fluid and traffic immune cells, but are also utilised by tumours to metastasise to lymph nodes and secondary sites in cancer. We characterise molecular mechanisms that control lymphangiogenesis using model organisms (zebrafish and mice). We particularly use single cell genomics coupled with systems biology approaches, large scale genetic screens, CRISPR genome editing and transgenesis to characterise molecular mechanisms. Current work includes detailed genetic analysis of pathways including the CCBE1/VEGFC/VEGFR3 pathway, the Hippo pathway, the role of alternative pre-mRNA splicing and the role of the cell fate regulator Prox1.

Genetics of blood brain barrier formation in zebrafish

The blood brain barrier (BBB) separates the brain parenchyma from circulating blood and macromolecules, representing a significant challenge for delivery of therapeutics, and acting as a barrier in metastasis. The BBB is made up of tightly adherent vascular endothelial cells surrounded by essential mural cell lineages, such as pericytes. How the different cells at the BBB develop, interact and function remains far from understood. We are using single cell genomics and a forward genetic screen in zebrafish to discover new genes and molecular mechanisms that control the maturation and function of pericytes and BBB endothelial cells. This work aims to identify new molecular mechanisms and future therapeutic opportunities.

Live imaging cell signalling and molecular dynamics in vascular development

Directly observing key molecules and pathways as they act to control tissue formation, or as they drive pathological phenotypes, is a major challenge in cell biology. We are utilising biosensors that report signalling events and multiple imaging modalities to directly observe real-time molecular functions in cells in normal development and disease models in vivo. This work has uncovered unexpected mechanisms in angiogenesis during wound repair and allowed us to study cell-cell adhesion mechanics in developing tissues. We are currently diversifying our vascular biosensor tools and applying new imaging approaches to observe dynamic changes in individual molecules of interest.


LAB MEMBERS
Baltaci, Oguzhan Post-Doc Mason, Elizabeth Post-Doc Okuda, Kazuhide Shaun Post-Doc
Brown, Ryan Graduate Student Kobayashi, Sakurako Graduate Student Usseglio Gaudi, Andrea Graduate Student
Yu, Hujun Graduate Student Chen, Tyrone Research Staff Dudczig, Stefanie Research Staff
Paterson, Scott Research Staff Rondon Galeano, Maria Cristina Research Staff


ZEBRAFISH PUBLICATIONS OF LAB MEMBERS
Ng, M.F., Da Silva Viana, J., Tan, P.J., Britto, D.D., Choi, S.B., Kobayashi, S., Samat, N., Song, D.S.S., Ogawa, S., Parhar, I.S., Astin, J.W., Hogan, B.M., Patel, V., Okuda, K.S. (2024) Canthin-6-One Inhibits Developmental and Tumour-Associated Angiogenesis in Zebrafish. Pharmaceuticals (Basel, Switzerland). 17(1):
Friedman, C.E., Cheetham, S.W., Negi, S., Mills, R.J., Ogawa, M., Redd, M.A., Chiu, H.S., Shen, S., Sun, Y., Mizikovsky, D., Bouveret, R., Chen, X., Voges, H.K., Paterson, S., De Angelis, J.E., Andersen, S.B., Cao, Y., Wu, Y., Jafrani, Y.M.A., Yoon, S., Faulkner, G.J., Smith, K.A., Porrello, E., Harvey, R.P., Hogan, B.M., Nguyen, Q., Zeng, J., Kikuchi, K., Hudson, J.E., Palpant, N.J. (2023) HOPX-associated molecular programs control cardiomyocyte cell states underpinning cardiac structure and function. Developmental Cell. 59(1):91-107.e6
Overman, J., Fontaine, F., Moustaqil, M., Mittal, D., Sierecki, E., Sacilotto, N., Zuegg, J., Robertson, A.A.B., Holmes, K., Salim, A.A., Mamidyala, S., Butler, M.S., Robinson, A.S., Lesieur, E., Johnston, W., Alexandrov, K., Black, B.L., Hogan, B.M., De Val, S., Capon, R.J., Carroll, J.S., Bailey, T.L., Koopman, P., Jauch, R., Cooper, M.A., Gambin, Y., Francois, M. (2023) Correction: Pharmacological targeting of the transcription factor SOX18 delays breast cancer in mice. eLIFE. 12
Leong, S.W., Wang, J., Okuda, K.S., Su, Q., Zhang, Y., Abas, F., Chia, S.L., Yusoff, K. (2023) Discovery of a novel dual functional phenylpyrazole-styryl hybrid that induces apoptotic and autophagic cell death in bladder cancer cells. European Journal of Medicinal Chemistry. 254:115335115335
Grimm, L., Mason, E., Yu, H., Dudczig, S., Panara, V., Chen, T., Bower, N.I., Paterson, S., Rondon Galeano, M., Kobayashi, S., Senabouth, A., Lagendijk, A.K., Powell, J., Smith, K.A., Okuda, K.S., Koltowska, K., Hogan, B.M. (2023) Single-cell analysis of lymphatic endothelial cell fate specification and differentiation during zebrafish development. The EMBO journal. 42(11):e112590
Morgan, K.J., Doggett, K., Geng, F., Mieruszynski, S., Whitehead, L., Smith, K.A., Hogan, B.M., Simons, C., Baillie, G.J., Molania, R., Papenfuss, A.T., Hall, T.E., Ober, E.A., Stainier, D.Y.R., Gong, Z., Heath, J.K. (2023) ahctf1 and kras mutations combine to amplify oncogenic stress and restrict liver overgrowth in a zebrafish model of hepatocellular carcinoma. eLIFE. 12:
Chau, T.C.Y., Keyser, M.S., Da Silva, J.A., Morris, E.K., Yordanov, T.E., Duscyz, K.P., Paterson, S., Yap, A.S., Hogan, B.M., Lagendijk, A.K. (2022) Dynamically regulated focal adhesions coordinate endothelial cell remodelling in developing vasculature. Development (Cambridge, England). 149(23):
Britto, D.D., He, J., Misa, J.P., Chen, W., Kakadia, P.M., Grimm, L., Herbert, C.D., Crosier, K.E., Crosier, P.S., Bohlander, S.K., Hogan, B.M., Hall, C.J., Torres-Vázquez, J., Astin, J.W. (2022) Plexind1 negatively regulates zebrafish lymphatic development. Development (Cambridge, England). 149(21)
Arnold, H., Panara, V., Hußmann, M., Filipek-Gorniok, B., Skoczylas, R., Ranefall, P., Gloger, M., Allalou, A., Hogan, B.M., Schulte-Merker, S., Koltowska, K. (2022) mafba and mafbb differentially regulate lymphatic endothelial cell migration in topographically distinct manners. Cell Reports. 39:110982
Gibson, J.F., Bojarczuk, A., Evans, R.J., Kamuyango, A.A., Hotham, R., Lagendijk, A.K., Hogan, B.M., Ingham, P.W., Renshaw, S.A., Johnston, S.A. (2022) Blood vessel occlusion by Cryptococcus neoformans is a mechanism for haemorrhagic dissemination of infection. PLoS pathogens. 18:e1010389
Koltowska, K., Okuda, K.S., Gloger, M., Rondon-Galeano, M., Mason, E., Xuan, J., Dudczig, S., Chen, H., Arnold, H., Skoczylas, R., Bower, N.I., Paterson, S., Lagendijk, A.K., Baillie, G.J., Leshchiner, I., Simons, C., Smith, K.A., Goessling, W., Heath, J.K., Pearson, R.B., Sanij, E., Schulte-Merker, S., Hogan, B.M. (2021) The RNA helicase Ddx21 controls Vegfc-driven developmental lymphangiogenesis by balancing endothelial cell ribosome biogenesis and p53 function. Nature cell biology. 23:1136-1147
Okuda, K.S., Ng, M.F., Ruslan, N.F., Bower, N.I., Song, D.S.S., Chen, H., Baek, S., Crosier, P.S., Koltowska, K., Astin, J.W., Tan, P.J., Hogan, B.M., Patel, V. (2021) 3,4-Difluorobenzocurcumin Inhibits Vegfc-Vegfr3-Erk Signalling to Block Developmental Lymphangiogenesis in Zebrafish. Pharmaceuticals (Basel, Switzerland). 14(7):
Wood, A.J., Lin, C.H., Li, M., Nishtala, K., Alaei, S., Rossello, F., Sonntag, C., Hersey, L., Miles, L.B., Krisp, C., Dudczig, S., Fulcher, A.J., Gibertini, S., Conroy, P.J., Siegel, A., Mora, M., Jusuf, P., Packer, N.H., Currie, P.D. (2021) FKRP-dependent glycosylation of fibronectin regulates muscle pathology in muscular dystrophy. Nature communications. 12:2951
Okuda, K.S., Keyser, M.S., Gurevich, D.B., Sturtzel, C., Mason, E.A., Paterson, S., Chen, H., Scott, M., Condon, N.D., Martin, P., Distel, M., Hogan, B.M. (2021) Live-imaging of endothelial Erk activity reveals dynamic and sequential signalling events during regenerative angiogenesis. eLIFE. 10:
Brandli, A., Dudczig, S., Currie, P.D., Jusuf, P.R. (2021) Photoreceptor ablation following ATP induced injury triggers Müller glia driven regeneration in zebrafish. Experimental Eye Research. 207:108569
Koopman, C.D., De Angelis, J., Iyer, S.P., Verkerk, A.O., Da Silva, J., Berecki, G., Jeanes, A., Baillie, G.J., Paterson, S., Uribe, V., Ehrlich, O.V., Robinson, S.D., Garric, L., Petrou, S., Simons, C., Vetter, I., Hogan, B.M., de Boer, T.P., Bakkers, J., Smith, K.A. (2021) The zebrafish grime mutant uncovers an evolutionarily conserved role for Tmem161b in the control of cardiac rhythm. Proceedings of the National Academy of Sciences of the United States of America. 118(9):
Xie, J., Jusuf, P.R., Bui, B.V., Dudczig, S., Sztal, T.E., Goodbourn, P.T. (2021) Altered Visual Function in a Larval Zebrafish Knockout of Neurodevelopmental Risk Gene pdzk1. Investigative ophthalmology & visual science. 62:29
Duszyc, K., Gomez, G.A., Lagendijk, A.K., Yau, M.K., Nanavati, B.N., Gliddon, B.L., Hall, T.E., Verma, S., Hogan, B.M., Pitson, S.M., Fairlie, D.P., Parton, R.G., Yap, A.S. (2021) Mechanotransduction activates RhoA in the neighbors of apoptotic epithelial cells to engage apical extrusion. Current biology : CB. 31(6):1326-1336.e5
Wong, E.S., Zheng, D., Tan, S.Z., Bower, N.L., Garside, V., Vanwalleghem, G., Gaiti, F., Scott, E., Hogan, B.M., Kikuchi, K., McGlinn, E., Francois, M., Degnan, B.M. (2020) Deep conservation of the enhancer regulatory code in animals. Science (New York, N.Y.). 370(6517):
Chaudhury, S., Okuda, K.S., Koltowska, K., Lagendijk, A.K., Paterson, S., Baillie, G.J., Simons, C., Smith, K.A., Hogan, B.M., Bower, N.I. (2020) Localised Collagen2a1 secretion supports lymphatic endothelial cell migration in the zebrafish embryo. Development (Cambridge, England). 147(18):
Okuda, K.S., Hogan, B.M. (2020) Endothelial Cell Dynamics in Vascular Development: Insights From Live-Imaging in Zebrafish. Frontiers in Physiology. 11:842
Rondon-Galeano, M., Skoczylas, R., Bower, N.I., Simons, C., Gordon, E., Francois, M., Koltowska, K., Hogan, B.M. (2020) MAFB modulates the maturation of lymphatic vascular networks in mice. Developmental Dynamics : an official publication of the American Association of Anatomists. 249(10):1201-1216
Vivien, C.J., Pichol-Thievend, C., Sim, C.B., Smith, J.B., Bower, N.I., Hogan, B.M., Hudson, J.E., Francois, M., Porrello, E.R. (2019) Vegfc/d-dependent regulation of the lymphatic vasculature during cardiac regeneration is influenced by injury context. NPJ Regenerative medicine. 4:18
Vogrin, A.J., Bower, N.I., Gunzburg, M.J., Roufail, S., Okuda, K.S., Paterson, S., Headey, S.J., Stacker, S.A., Hogan, B.M., Achen, M.G. (2019) Evolutionary Differences in the Vegf/Vegfr Code Reveal Organotypic Roles for the Endothelial Cell Receptor Kdr in Developmental Lymphangiogenesis. Cell Reports. 28:2023-2036.e4
Ehrlich, O., Karamalakis, A., Krylov, A.J., Dudczig, S., Hassell, K.L., Jusuf, P.R. (2019) Clove Oil and AQUI-S Efficacy for Zebrafish Embryo, Larva, and Adult Anesthesia. Zebrafish. 16(5):451-459
Grimm, L., Nakajima, H., Chaudhury, S., Bower, N.I., Okuda, K.S., Cox, A.G., Harvey, N.L., Koltowska, K., Mochizuki, N., Hogan, B.M. (2019) Yap1 promotes sprouting and proliferation of lymphatic progenitors downstream of Vegfc in the zebrafish trunk. eLIFE. 8:
Baek, S., Oh, T.G., Secker, G., Sutton, D.L., Okuda, K.S., Paterson, S., Bower, N.I., Toubia, J., Koltowska, K., Capon, S.J., Baillie, G.J., Simons, C., Muscat, G.E.O., Lagendijk, A.K., Smith, K.A., Harvey, N.L., Hogan, B.M. (2019) The Alternative Splicing Regulator Nova2 Constrains Vascular Erk Signaling to Limit Specification of the Lymphatic Lineage. Developmental Cell. 49:279-292.e5
Eng, T.C., Chen, W., Okuda, K.S., Misa, J.P., Padberg, Y., Crosier, K.E., Crosier, P.S., Hall, C.J., Schulte-Merker, S., Hogan, B.M., Astin, J.W. (2019) Zebrafish facial lymphatics develop through sequential addition of venous and non-venous progenitors. EMBO reports. 20(5):
Grassini, D.R., Da Silva, J., Hall, T.E., Baillie, G.J., Simons, C., Parton, R.G., Hogan, B.M., Smith, K.A. (2019) Myosin Vb is required for correct trafficking of N-cadherin and cardiac chamber ballooning. Developmental Dynamics : an official publication of the American Association of Anatomists. 248(4):284-295
Ando, K., Wang, W., Peng, D., Chiba, A., Lagendijk, A., Barske, L., Crump, J.G., Stainier, D.Y.R., Lendahl, U., Koltowska, K., Hogan, B.M., Fukuhara, S., Mochizuki, N., Betsholtz, C. (2019) Peri-arterial specification of vascular mural cells from naïve mesenchyme requires Notch signaling. Development (Cambridge, England). 146(2):
Tan, P. J., Lau, B. F., Krishnasamy, G., Ng, M. F., Husin, L. S., Ruslan, N., Song, D. S.S., Velaithan, V., Okuda, K. S., and Patel, V. (2018) Zebrafish embryonic development-interfering macrolides from Streptomyces californicus impact growth and mitochondrial function in human colorectal cancer cells. Process Biochemistry. 74:164-174
Paatero, I., Sauteur, L., Lee, M., Lagendijk, A.K., Heutschi, D., Wiesner, C., Guzmán, C., Bieli, D., Hogan, B.M., Affolter, M., Belting, H.G. (2018) Junction-based lamellipodia drive endothelial cell rearrangements in vivo via a VE-cadherin-F-actin based oscillatory cell-cell interaction. Nature communications. 9:3545
Samat, N., Ng, M.F., Ruslan, N.F., Okuda, K.S., Tan, P.J., Patel, V. (2018) Interference Potential of Tannins and Chlorophylls in Zebrafish Phenotypic-Based Assays. Assay and drug development technologies. 16(7):408-419
Grassini, D.R., Lagendijk, A.K., De Angelis, J.E., Da Silva, J., Jeanes, A., Zettler, N., Bower, N.I., Hogan, B.M., Smith, K.A. (2018) Nppa and Nppb act redundantly during zebrafish cardiac development to confine AVC marker expression and reduce cardiac jelly volume. Development (Cambridge, England). 145(12)
Bower, N.I., Hogan, B.M. (2018) Brain drains: new insights into brain clearance pathways from lymphatic biology. Journal of molecular medicine (Berlin, Germany). 96(5):383-390
Okuda, K.S., Baek, S., Hogan, B.M. (2018) Visualization and Tools for Analysis of Zebrafish Lymphatic Development. Methods in molecular biology (Clifton, N.J.). 1846:55-70
Lagendijk, A.K., Gomez, G.A., Baek, S., Hesselson, D., Hughes, W.E., Paterson, S., Conway, D.E., Belting, H.G., Affolter, M., Smith, K.A., Schwartz, M.A., Yap, A.S., Hogan, B.M. (2017) Live imaging molecular changes in junctional tension upon VE-cadherin in zebrafish. Nature communications. 8:1402
Dudczig, S., Currie, P.D., Jusuf, P.R. (2017) Developmental and adult characterization of secretagogin expressing amacrine cells in zebrafish retina. PLoS One. 12:e0185107
Hogan, B.M., Schulte-Merker, S. (2017) How to Plumb a Pisces: Understanding Vascular Development and Disease Using Zebrafish Embryos. Developmental Cell. 42:567-583
Chiang, I.K., Fritzsche, M., Pichol-Thievend, C., Neal, A., Holmes, K., Lagendijk, A., Overman, J., D'Angelo, D., Omini, A., Hermkens, D., Lesieur, E., Liu, K., Ratnayaka, I., Corada, M., Bou-Gharios, G., Carroll, J., Dejana, E., Schulte-Merker, S., Hogan, B., Beltrame, M., De Val, S., Francois, M. (2017) SoxF factors induce Notch1 expression via direct transcriptional regulation during early arterial development. Development (Cambridge, England). 144(14):2629-2639
Bower, N.I., Koltowska, K., Pichol-Thievend, C., Virshup, I., Paterson, S., Lagendijk, A.K., Wang, W., Lindsey, B.W., Bent, S.J., Baek, S., Rondon-Galeano, M., Hurley, D.G., Mochizuki, N., Simons, C., Francois, M., Wells, C.A., Kaslin, J., Hogan, B.M. (2017) Mural lymphatic endothelial cells regulate meningeal angiogenesis in the zebrafish. Nature Neuroscience. 20(6):774-783
Velaithan, V., Okuda, K.S., Ng, M.F., Samat, N., Leong, S.W., Faudzi, S.M., Abas, F., Shaari, K., Cheong, S.C., Tan, P.J., Patel, V. (2017) Zebrafish phenotypic screen identifies novel notch antagonists. Investigational new drugs. 35(2):166-179
Velaithan, V., Okuda, K.S., Ng, M.F., Samat, N., Leong, S.W., Faudzi, S.M., Abas, F., Shaari, K., Cheong, S.C., Tan, P.J., Patel, V. (2017) Erratum to: Zebrafish phenotypic screen identifies novel Notch antagonists. Investigational new drugs.
Dudczig, S., Currie, P.D., Poggi, L., Jusuf, P.R. (2017) In Vivo Imaging of Transgenic Gene Expression in Individual Retinal Progenitors in Chimeric Zebrafish Embryos to Study Cell Nonautonomous Influences. Journal of visualized experiments : JoVE. (121)
Bower, N.I., Vogrin, A.J., Le Guen, L., Chen, H., Stacker, S.A., Achen, M.G., Hogan, B.M. (2017) Vegfd modulates both angiogenesis and lymphangiogenesis during zebrafish embryonic development. Development (Cambridge, England). 144(3):507-518
Dalton, J.P., Uy, B., Okuda, K.S., Hall, C.J., Denny, W.A., Crosier, P.S., Swift, S., Wiles, S. (2017) Screening of anti-mycobacterial compounds in a naturally infected zebrafish larvae model. The Journal of antimicrobial chemotherapy. 72(2):421-427
Overman, J., Fontaine, F., Moustaqil, M., Mittal, D., Sierecki, E., Sacilotto, N., Zuegg, J., Robertson, A.A., Holmes, K., Salim, A.A., Mamidyala, S., Butler, M.S., Robinson, A.S., Lesieur, E., Johnston, W., Alexandrov, K., Black, B.L., Hogan, B.M., Val, S., Capon, R.J., Carroll, J.S., Bailey, T.L., Koopman, P., Jauch, R., Smyth, M.J., Cooper, M.A., Gambin, Y., Francois, M. (2017) Pharmacological targeting of the transcription factor SOX18 delays breast cancer in mice. eLIFE. 6
De Angelis, J.E., Lagendijk, A.K., Chen, H., Tromp, A., Bower, N.I., Tunny, K.A., Brooks, A.J., Bakkers, J., Francois, M., Yap, A.S., Simons, C., Wicking, C., Hogan, B.M., Smith, K.A. (2017) Tmem2 Regulates Embryonic Vegf Signaling by Controlling Hyaluronic Acid Turnover. Developmental Cell. 40:123-136
Capon, S.J., Baillie, G.J., Bower, N.I., da Silva, J.A., Paterson, S., Hogan, B.M., Simons, C., Smith, K.A. (2017) Utilising polymorphisms to achieve allele-specific genome editing in zebrafish. Biology Open. 6(1):125-131
Crawford, J., Bower, N.I., Hogan, B.M., Taft, R.J., Gabbett, M.T., McGaughran, J., Simons, C. (2016) Expanding the genotypic spectrum of CCBE1 mutations in Hennekam syndrome. American journal of medical genetics. Part A. 170(10):2694-7
Ng Chi Kei, J., Dudczig, S., Currie, P.D., Jusuf, P.R. (2016) Feedback from each retinal neuron population drives expression of subsequent fate determinant genes without influencing the cell cycle exit timing. The Journal of comparative neurology. 524(13):2553-66
Okuda, K.S., Lee, H.M., Velaithan, V., Ng, M.F., Patel, V. (2016) Utilizing zebrafish to identify anti-(lymph)angiogenic compounds for cancer treatment: promise and future challenges. Microcirculation (New York, N.Y. : 1994). 23(6):389-405
Okuda, K.S., Tan, P.J., Patel, V. (2016) Sprouting Buds of Zebrafish Research in Malaysia: First Malaysia Zebrafish Disease Model Workshop. Zebrafish. 13(2):138-41
Koenig, A.L., Baltrunaite, K., Bower, N.I., Rossi, A., Stainier, D.Y., Hogan, B.M., Sumanas, S. (2016) Vegfa signaling promotes zebrafish intestinal vasculature development through endothelial cell migration from the posterior cardinal vein. Developmental Biology. 411(1):115-27
Koltowska, K., Lagendijk, A.K., Pichol-Thievend, C., Fischer, J.C., Francois, M., Ober, E.A., Yap, A.S., Hogan, B.M. (2015) Vegfc Regulates Bipotential Precursor Division and Prox1 Expression to Promote Lymphatic Identity in Zebrafish. Cell Reports. 13:1828-41
Boije, H., Rulands, S., Dudczig, S., Simons, B.D., Harris, W.A. (2015) The Independent Probabilistic Firing of Transcription Factors: A Paradigm for Clonal Variability in the Zebrafish Retina. Developmental Cell. 34(5):532-43
Okuda, K.S., Misa, J.P., Oehlers, S.H., Hall, C.J., Ellett, F., Alasmari, S., Lieschke, G.J., Crosier, K.E., Crosier, P.S., Astin, J.W. (2015) A zebrafish model of inflammatory lymphangiogenesis. Biology Open. 4(10):1270-80
Koltowska, K., Paterson, S., Bower, N.I., Baillie, G.J., Lagendijk, A.K., Astin, J.W., Chen, H., Francois, M., Crosier, P.S., Taft, R.J., Simons, C., Smith, K.A., Hogan, B.M. (2015) mafba is a downstream transcriptional effector of Vegfc signaling essential for embryonic lymphangiogenesis in zebrafish. Genes & Development. 29:1618-30
Jeffery, J., Neyt, C., Moore, W., Paterson, S., Bower, N.I., Chenevix-Trench, G., Verkade, H., Hogan, B.M., Khanna, K.K. (2015) Cep55 regulates embryonic growth and development by promoting Akt stability in zebrafish. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 29(5):1999-2009
Oehlers, S.H., Cronan, M.R., Scott, N.R., Thomas, M.I., Okuda, K.S., Walton, E.M., Beerman, R.W., Crosier, P.S., Tobin, D.M. (2015) Interception of host angiogenic signalling limits mycobacterial growth. Nature. 517(7536):612-5
Coxam, B., Neyt, C., Grassini, D.R., Le Guen, L., Smith, K.A., Schulte-Merker, S., Hogan, B.M. (2015) carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase (cad) regulates Notch signaling and vascular development in zebrafish. Developmental Dynamics : an official publication of the American Association of Anatomists. 244(1):1-9
Astin, J.W., Haggerty, M.J., Okuda, K.S., Le Guen, L., Misa, J.P., Tromp, A., Hogan, B.M., Crosier, K.E., Crosier, P.S. (2014) Vegfd can compensate for loss of Vegfc in zebrafish facial lymphatic sprouting. Development (Cambridge, England). 141(13):2680-90
Coxam, B., Sabine, A., Bower, N.I., Smith, K.A., Pichol-Thievend, C., Skoczylas, R., Astin, J.W., Frampton, E., Jaquet, M., Crosier, P.S., Parton, R.G., Harvey, N.L., Petrova, T.V., Schulte-Merker, S., Francois, M., Hogan, B.M. (2014) Pkd1 Regulates Lymphatic Vascular Morphogenesis during Development. Cell Reports. 7:623-33
Kartopawiro, J., Bower, N.I., Karnezis, T., Kazenwadel, J., Betterman, K.L., Lesieur, E., Koltowska, K., Astin, J., Crosier, P., Vermeren, S., Achen, M.G., Stacker, S.A., Smith, K.A., Harvey, N.L., François, M., and Hogan, B.M. (2014) Arap3 is dysregulated in a mouse model of hypotrichosis-lymphedema-telangiectasia and regulates lymphatic vascular development. Human molecular genetics. 23(5):1286-97
Le Guen, L., Karpanen, T., Schulte, D., Harris, N.C., Koltowska, K., Roukens, G., Bower, N.I., van Impel, A., Stacker, S.A., Achen, M.G., Schulte-Merker, S., and Hogan, B.M. (2014) Ccbe1 regulates Vegfc-mediated induction of Vegfr3 signaling during embryonic lymphangiogenesis. Development (Cambridge, England). 141(6):1239-1249
Duong, T., Koltowska, K., Pichol-Thievend, C., Le Guen, L., Fontaine, F., Smith, K.A., Truong, V., Skoczylas, R., Stacker, S.A., Achen, M.G., Koopman, P., Hogan, B.M., and Francois, M. (2014) VEGFD regulates blood vascular development by modulating SOX18 activity. Blood. 123(7):1102-12
Chang, G.H., Lay, A.J., Ting, K.K., Zhao, Y., Coleman, P.R., Powter, E.E., Formaz-Preston, A., Jolly, C.J., Bower, N.I., Hogan, B.M., Rinkwitz, S., Becker, T.S., Vadas, M.A., Gamble, J.R. (2014) ARHGAP18: an endogenous inhibitor of angiogenesis, limiting tip formation and stabilizing junctions. Small GTPases. 5(3):1-15
Lagendijk, A.K., Yap, A.S., Hogan, B.M. (2014) Endothelial cell-cell adhesion during zebrafish vascular development. Cell adhesion & migration. 8(2):136-145
Keightley, M.C., Crowhurst, M.O., Layton, J.E., Beilharz, T., Markmiller, S., Varma, S., Hogan, B.M., de Jong-Curtain, T.A., Heath, J.K., and Lieschke, G.J. (2013) In vivo mutation of pre-mRNA processing factor 8 (Prpf8) affects transcript splicing, cell survival and myeloid differentiation. FEBS letters. 587(14):2150-2157
Cermenati, S., Moleri, S., Neyt, C., Bresciani, E., Carra, S., Grassini, D.R., Omini, A., Goi, M., Cotelli, F., François, M., Hogan, B.M., and Beltrame, M. (2013) Sox18 Genetically Interacts With VegfC to Regulate Lymphangiogenesis in Zebrafish. Arterioscler. Thromb. Vasc. Biol.. 33(6):1238-47
Oehlers, S.H., Flores, M.V., Hall, C.J., Okuda, K.S., Sison, J.O., Crosier, K.E., and Crosier, P.S. (2013) Chemically Induced Intestinal Damage Models in Zebrafish Larvae. Zebrafish. 10(2):184-93
Koltowska, K., Betterman, K.L., Harvey, N.L., and Hogan, B.M. (2013) Getting out and about: the emergence and morphogenesis of the vertebrate lymphatic vasculature. Development (Cambridge, England). 140(9):1857-1870
Kizil, C., Kyritsis, N., Dudczig, S., Kroehne, V., Freudenreich, D., Kaslin, J., and Brand, M. (2012) Regenerative Neurogenesis from Neural Progenitor Cells Requires Injury-Induced Expression of Gata3. Developmental Cell. 23(6):1230-1237
Macheda, M.L., Sun, W.W., Kugathasan, K., Hogan, B.M., Bower, N.I., Halford, M.M., Zhang, Y.F., Jacques, B.E., Lieschke, G.J., Dabdoub, A., and Stacker, S.A. (2012) The Wnt Receptor Ryk Plays a Role in Mammalian Planar Cell Polarity Signaling. The Journal of biological chemistry. 287(35):29312-29323
Kizil, C., Dudczig, S., Kyritsis, N., Machate, A., Blaesche, J., Kroehne, V., and Brand, M. (2012) The chemokine receptor cxcr5 regulates the regenerative neurogenesis response in the adult zebrafish brain. Neural Development. 7(1):27
Okuda, K.S., Astin, J.W., Misa, J.P., Flores, M.V., Crosier, K.E., and Crosier, P.S. (2012) lyve1 expression reveals novel lymphatic vessels and new mechanisms for lymphatic vessel development in zebrafish. Development (Cambridge, England). 139(13):2381-2391
Smith, K.A., Lagendijk, A.K., Courtney, A.D., Chen, H., Paterson, S., Hogan, B.M., Wicking, C., and Bakkers, J. (2011) Transmembrane protein 2 (Tmem2) is required to regionally restrict atrioventricular canal boundary and endocardial cushion development. Development (Cambridge, England). 138(19):4193-4198
Oehlers, S.H., Flores, M.V., Okuda, K.S., Hall, C.J., Crosier, K.E., and Crosier, P.S. (2011) A chemical enterocolitis model in zebrafish larvae that is dependent on microbiota and responsive to pharmacological agents. Developmental Dynamics : an official publication of the American Association of Anatomists. 240(1):288-298
Tao, S., Witte, M., Bryson-Richardson, R.J., Currie, P.D., Hogan, B.M., and Schulte-Merker, S. (2011) Zebrafish prox1b Mutants Develop a Lymphatic Vasculature, and prox1b Does Not Specifically Mark Lymphatic Endothelial Cells. PLoS One. 6(12):e28934
Geudens, I., Herpers, R., Hermans, K., Segura, I., Ruiz de Almodovar, C., Bussmann, J., De Smet, F., Vandevelde, W., Hogan, B.M., Siekmann, A., Claes, F., Moore, J.C., Pistocchi, A.S., Loges, S., Mazzone, M., Mariggi, G., Bruyere, F., Cotelli, F., Kerjaschki, D., Noel, A., Foidart, J.M., Gerhardt, H., Ny, A., Langenberg, T., Lawson, N.D., Duckers, H.J., Schulte-Merker, S., Carmeliet, P., and Dewerchin, M. (2010) Role of delta-like-4/Notch in the formation and wiring of the lymphatic network in zebrafish. Arterioscler. Thromb. Vasc. Biol.. 30(9):1695-1702
Alders, M., Hogan, B.M., Gjini, E., Salehi, F., Al-Gazali, L., Hennekam, E.A., Holmberg, E.E., Mannens, M.M., Mulder, M.F., Offerhaus, G.J., Prescott, T.E., Schroor, E.J., Verheij, J.B., Witte, M., Zwijnenburg, P.J., Vikkula, M., Schulte-Merker, S., and Hennekam, R.C. (2009) Mutations in CCBE1 cause generalized lymph vessel dysplasia in humans. Nature Genetics. 41(12):1272-1274
Hogan, B.M., Herpers, R., Witte, M., Heloterä, H., Alitalo, K., Duckers, H.J., and Schulte-Merker, S. (2009) Vegfc/Flt4 signalling is suppressed by Dll4 in developing zebrafish intersegmental arteries. Development (Cambridge, England). 136(23):4001-4009
Voss, K., Stahl, S., Hogan, B.M., Reinders, J., Schleider, E., Schulte-Merker, S., and Felbor, U. (2009) Functional analyses of human and zebrafish 18-amino acid in-frame deletion pave the way for domain mapping of the cerebral cavernous malformation 3 protein. Human Mutation. 30(6):1003-1011
Hogan, B.M., Bos, F.L., Bussmann, J., Witte, M., Chi, N.C., Duckers, H.J., and Schulte-Merker, S. (2009) ccbe1 is required for embryonic lymphangiogenesis and venous sprouting. Nature Genetics. 41(4):396-398
Soroldoni, D., Hogan, B.M., and Oates, A.C. (2009) Simple and efficient transgenesis with meganuclease constructs in zebrafish. Methods in molecular biology (Clifton, N.J.). 546:117-130
Hogan, B.M., Bussmann, J., Wolburg, H., and Schulte-Merker, S. (2008) ccm1 cell autonomously regulates endothelial cellular morphogenesis and vascular tubulogenesis in zebrafish. Human molecular genetics. 17(16):2424-2432
Hogan, B.M., Verkade, H., Lieschke, G.J., and Heath, J.K. (2008) Manipulation of gene expression during zebrafish embryonic development using transient approaches. Methods in molecular biology (Clifton, N.J.). 469:273-300
Dworkin, S., Heath, J.K., Dejong-Curtain, T.A., Hogan, B.M., Lieschke, G.J., Malaterre, J., Ramsay, R.G., and Mantamadiotis, T. (2007) CREB activity modulates neural cell proliferation, midbrain-hindbrain organization and patterning in zebrafish. Developmental Biology. 307(1):127-141
Hogan, B.M., Pase, L., Hall, N.E., and Lieschke, G.J. (2006) Characterisation of duplicate zinc finger like 2 erythroid precursor genes in zebrafish. Development genes and evolution. 216(9):523-529
Hogan, B.M., Layton, J.E., Pyati, U.J., Nutt, S.L., Hayman, J.W., Varma, S., Heath, J.K., Kimelman, D., and Lieschke, G.J. (2006) Specification of the Primitive Myeloid Precursor Pool Requires Signaling through Alk8 in Zebrafish. Current biology : CB. 16(5):506-511
Hogan, B.M., Danks, J.A., Layton, J.E., Hall, N.E., Heath, J.K., and Lieschke, G.J. (2005) Duplicate zebrafish pth genes are expressed along the lateral line and in the central nervous system during embryogenesis. Endocrinology. 146(2):547-551
Hogan, B.M., Hunter, M.P., Oates, A.C., Crowhurst, M.O., Hall, N.E., Heath, J.K., Prince, V.E., and Lieschke, G.J. (2004) Zebrafish gcm2 is required for gill filament budding from pharyngeal ectoderm. Developmental Biology. 276(2):508-522
Mawdsley, D.J., Cooper, H.M., Hogan, B.M., Cody, S.H., Lieschke, G.J., and Heath, J.K. (2004) The Netrin receptor Neogenin is required for neural tube formation and somitogenesis in zebrafish. Developmental Biology. 269(1):302-315
Hogan, B.M. (2004) Characterisation of candidate haemopoietic regulators by genetic approaches in zebrafish. Ph.D. Thesis. :286p