ZFIN ID: ZDB-PERS-970213-1
Krumlauf, Robb
Email: rek@stowers.org
URL: http://www.stowers.org
Affiliation: Krumlauf Lab
Address: Stowers Institute for Medical Research 1000 East 50th Street Kansas City, MO 64110 USA
Country: United States
Phone: (816) 926-4051
Fax:
ORCID ID: 0000-0001-9102-7927


BIOGRAPHY AND RESEARCH INTERESTS
I am deeply interested in understanding the regulatory information and associated molecular mechanisms encoded in vertebrate genomes which guide the formation and elaboration of the basic body plan. My research has been aimed at dissecting regulatory circuits which control how organs and tissues are generated along embryonic axes form from similar cellular building blocks. A deep understanding of such processes is critical to understand morphogenesis. While there is amazing similarity in many processes between vertebrates how variations in their regulation leads to diversity in evolution is poorly understood. My group has use head development and Hox genes as model systems to understand patterning mechanisms and regulatory networks in hindbrain segmentation and craniofacial development. Hox genes play critical roles in regulating regional diversity in many tissues so our goal is to build a basis for comparing regulatory pathways that pattern many elements of the basic body plan in development, disease and evolution.

The vertebrate hindbrain and its relationship to head development is a good model system for understanding fundamental mechanisms of patterning and morphogenesis during development, disease and evolution. The hindbrain is a highly conserved complex co-ordination center in the vertebrate CNS. The formation of regional diversity in the hindbrain is achieved through a process of segmentation, which ultimately gives rise to well-defined regions of the adult brain. This segmental organization is critical for patterning of the cranial neural crest which generates most of the bone and connective tissues of head and facial structures. The Hox family of transcription factors is coupled to this process and provides a molecular framework for specifying the unique identities of hindbrain segments and facial structures. Because the segmental processes of head development are highly conserved among vertebrates, comparative studies between different species have greatly enhanced our ability to build a picture of the regulatory cascades that control early head development. Through comparative studies in lamprey, zebrafish and mice we are beginning to address the interesting question of when ordered domains of Hox expression were coupled to hindbrain segmentation in chordate origins and what signaling pathways/regulatory mechanisms were involved.

Biography: I received a BE in chemical engineering from Vanderbilt University (1970) and a PhD in developmental biology from Ohio State University (1979). I did postdoctoral research with Shirley Tilghman at the Fox Chase Cancer Center and established my group at England’s National Institute for Medical Research at Mill Hill, London, now a part of the Francis Crick Institute, where I became head of the Division of Developmental Neurobiology. I was the the Founding Scientific Director of the Stowers Institute (2000-2019) and am currently an Investigator and Scientific Director Emeritus of the Stowers Institute. I hold secondary faculty appointments at the University of Missouri at Kansas City Dental School and the University of Kansas Medical Center Department of Anatomy and Cell Biology.


PUBLICATIONS
Parker, H.J., De Kumar, B., Pushel, I., Bronner, M.E., Krumlauf, R. (2021) Analyses of lamprey meis genes reveals that conserved inputs from hox, Meis and Pbx proteins control their expression in the hindbrain and neural tube. Developmental Biology. 479:61-76
Prummel, K.D., Hess, C., Nieuwenhuize, S., Parker, H.J., Rogers, K.W., Kozmikova, I., Racioppi, C., Brombacher, E.C., Czarkwiani, A., Knapp, D., Burger, S., Chiavacci, E., Shah, G., Burger, A., Huisken, J., Yun, M.H., Christiaen, L., Kozmik, Z., Müller, P., Bronner, M., Krumlauf, R., Mosimann, C. (2019) A conserved regulatory program initiates lateral plate mesoderm emergence across chordates. Nature communications. 10:3857
Parker, H.J., De Kumar, B., Green, S.A., Prummel, K.D., Hess, C., Kaufman, C.K., Mosimann, C., Wiedemann, L.M., Bronner, M.E., Krumlauf, R. (2019) A Hox-TALE regulatory circuit for neural crest patterning is conserved across vertebrates. Nature communications. 10:1189
De Kumar, B., Parker, H.J., Paulson, A., Parrish, M.E., Zeitlinger, J., Krumlauf, R. (2017) Hoxa1 targets signaling pathways during neural differentiation of ES cells and mouse embryogenesis. Developmental Biology. 432(1):151-164
De Kumar, B., Parker, H.J., Paulson, A., Parrish, M.E., Pushel, I., Singh, N.P., Zhang, Y., Slaughter, B.D., Unruh, J.R., Florens, L., Zeitlinger, J., Krumlauf, R. (2017) HOXA1 and TALE proteins display cross-regulatory interactions and form a combinatorial binding code on HOXA1 targets. Genome research. 27(9):1501-1512
McEllin, J.A., Alexander, T.B., Tümpel, S., Wiedemann, L.M., Krumlauf, R. (2016) Analyses of fugu hoxa2 genes provide evidence for subfunctionalization of neural crest cell and rhombomere cis-regulatory modules during vertebrate evolution. Developmental Biology. 409(2):530-42
Parker, H.J., Bronner, M.E., Krumlauf, R. (2014) A Hox regulatory network of hindbrain segmentation is conserved to the base of vertebrates. Nature. 514(7523):490-3
Trainor, P.A. and Krumlauf, R. (2000) Patterning the cranial neural crest: hindbrain segmentation and Hox gene plasticity. Nature reviews. Neuroscience. 1(2):116-124
Manzanares, M., Trainor, P.A., Nonchev, S., Ariza-McNaughton, L., Brodie, J., Gould, A., Marshall, H., Morrison, A., Kwan, C.-T., Sham, M.-H., Wilkinson, D.G., and Krumlauf, R. (1999) The role of kreisler in segmentation during hindbrain development. Developmental Biology. 211(2):220-237
Pöpperl, H., Bienz, M., Studer, M., Chan, S., Aparicio, S., Brenner, S., Mann, R., and Krumlauf, R. (1995) Segmental expression of Hoxb-1 is controlled by a highly conserved autoregulatory loop dependent upon exd/pbx. Cell. 81:1031-1042
Graham, A., Papalopulu, N., Lorimer, J., McVey, J.H., Tuddenham, E.G.D., and Krumlauf, R. (1988) Characterization of a murine homeo box gene, Hox-2.6, related to the Drosophila Deformed gene.. Genes & Development. 2(11):1424-1438

NON-ZEBRAFISH PUBLICATIONS
Cambronero, F., et al (2019) Inter-rhombomeric interactions reveal roles for Fgf signaling in segmental regulation of EphA4 expression. Developmental Dynamics Epub ahead of print doi: 10.1002/dvdy.101
Prummel, K.D. et al (2019) A conserved regulatory program drives emergence of the lateral plate mesoderm. Nature Communications (In press)
Nolte, C., et al (2019) Hox genes: Downstream “effectors” of retinoic acid signaling in vertebrate embryogenesis. Genesis 57: doi: 10.1002/dvg.23306
Parker, H.J., et al (2019) An atlas of anterior hox gene expression in the embryonic sea lamprey head: hox-code evolution in vertebrates. Developmental Biology 453: 19-33,
Parker, H.J., et al (2019) A Hox-TALE regulatory circuit for neural crest patterning is conserved across vertebrates. Nature Communications 10: article 1189 doi: https://doi.org/10.1038/s41467-019-09197-8
Qian,P. et al (2018) Retinoid-sensitive epigenetic regulation of the Hoxb cluster maintains normal hematopoiesis and inhibits leukemogenesis. Cell Stem Cell 22: 740-754.
Parker, H.J., et al (2018) Coupling the roles of Hox genes to regulatory networks patterning cranial neural crest. Developmental Biology 444:Suppl 1, 67-78
Smith, J.J., et al (2018) The sea lamprey germline genome: evolution of form and function in vertebrate genomes. Nature Genetics 50: 270-277
De Kumar, B., et al (2017) Hoxa1 targets signaling pathways during neural differentiation of ES cells and mouse embryogenesis. Developmental Biology 432: 151-164.
De Kumar, B., et al (2017) Hoxa1 and TALE proteins display cross-regulatory interactions and form a combinatorial binding code on Hoxa1 targets. Genome Research, 27:1501-1512.
DeKumar, B., et al (2017). Dynamic regulation of Nanog and stem cell signaling pathways by Hoxa1 during early neuro-ectodermal differentiation of ES cells. PNAS 114: 5838-5845.
Ahn,Y., et al (2017) Multiple modes of Lrp4 function in modulation of Wnt/β-catenin signaling during tooth development. Development 144: 2824-2836.
Parker, H.J. and Krumlauf, R (2017) Segmental arithmetic: summing up the Hox gene regulatory network for hindbrain development in chordates. WIREs Developmental Biology, e286.
Parker, H., et al (2016) Evolution and diversification of the vertebrate Hox gene regulatory network for hindbrain development. Bioessays 38: 526-538. doi: 10.1002/bies.201600010.
De Kumar, B. and Krumlauf, R. (2016) HOXs and lincRNAs: Two sides of the same coin. Science Advances 2:e1501402.
De Kumar, B. et al (2015) Analysis of dynamic changes in retinoid induced transcription and epigenetic profiles of murine Hox clusters in ES cells. Genome Research 25:1229-1243.
Parker, H., et al (2014) A Hox regulatory network for hindbrain segmentation is conserved to the base of vertebrates. Nature. 514:490-493.
Ahn, Y., et al (2014) Long-range regulation by shared retinoic acid response elements modulates dynamic expression of posterior Hoxb genes in CNS development Developmental Biology 388:134-144.
Nolte, C., et al (2013) Shadow enhancers flanking the HoxB cluster direct dynamic Hox expression in early heart and endoderm development. Developmental Biology. 383:158-173.
Soshnikova, N., et al (2013) Duplications of Hox gene clusters and the emergence of vertebrates. Developmental Biology. 378:194–199.
Smith, J.J. et al (2013) The Lamprey Genome: Illuminating Vertebrate Origins Nature Genetics 45: 415–421.
Ahn, Y., et al (2013) Lrp4 and Wise interplay controls the formation and patterning of mammary and other skin appendage placodes by modulating Wnt signaling. Development 140: 583-593.
Natale,A., et al (2011) Evolution of anterior Hox regulatory elements among Chordates. BMC Evolutionary Biology 11:330-348.
Ahn, Y., et al (2010) Inhibition of Wnt signaling by Wise/Sostdc1 and negative feedback from Shh controls tooth number and patterning. Development 137:3221-3231.
Alexander, T., et al (2009) Hox genes and vertebrate segmentation. In: Annual Review of Cell and Developmental Biology 25: 431-456.
Tümpel, S., et al (2008) The Hox hindbrain regulatory network: A regulatory module embedded in the coding region of Hoxa2 controls expression in rhombomere 2. PNAS 105: 20077-20082.
Tümpel, S., et al (2006) Evolution of cis-elements in the differential expression of two Hoxa2 co-paralogous genes in pufferfish. PNAS 103: 5419-5424.
Serpente, P., et al (2005) Direct crossregulation between retinoic acid receptor β (RARand Hox genes during hindbrain segmentation. Development 132: 503-513.
Gavalas, A., et al (2003) Neuronal defects in the hindbrain of Hoxa1, Hoxb1 and Hoxb2 mutants reflect regulatory interactions among these Hox genes. Development, 130: 5663-5679.
Itasaki, N., et al (2003) Wise, a context dependent activator or inhibitor of Wnt signalling. Development, 130: 4295-4305. P
Bel-Vialar, S., et al (2002) Initiating Hox gene expression in the chick neural tube: Differential sensitivity to FGF and RA signalling defines two distinct groups of Hoxb genes. Development 129, 5103-5115.
Manzanares, M., et al (2002) Krox20 and kreisler cooperate in the transcriptional control of segmental expression of Hoxb3 in the developing hindbrain. EMBO J. 21, 365-376.
Trainor, P., et al (2002) Signalling between the hindbrain and paraxial tissues dictates neural crest migration pathways. Development, 129, 433-442.
Trainor, P., et al (2002) Role of the isthmus and FGFs in resolving the paradox of neural crest plasticity and prepatterning. Science, 295, 1288-129.
Manzanares, et al (2001) Independent regulation of initiation and maintenance phases of Hoxa3 expression in the vertebrate hindbrain involve auto and cross-regulatory mechanisms. Development, 128, 3595-3607.
Manzanares, M., et al (2000) Conservation and elaboration of Hox gene regulation during evolution of the vertebrate head. Nature 408, 854-857.
Bulman, M.P., et al (2000) Mutations in the human Delta homologue, DLL3, cause axial skeletal defects in spondylocostal dysostosis. Nature Genetics 24, 438-441.
Golding, J.P., et al (2000). Defects in pathfinding by cranial neural crest cells in mice lacking the neuregulin receptor ErbB4. Nature Cell Biology, 2, 103-109.
Trainor, P. and Krumlauf, R. (2000) Plasticity in mouse neural crest cells reveals a novel patterning role for cranial mesoderm. Nature Cell Biology, 2, 96-102.
Maconochie, M., et al (1999) Regulation of Hoxa2 in cranial neural crest cells involves members of the AP-2 family. Development, 126: 1483-1494.
Sharpe, J., et al (1999). Identification of Sonic hedgehog as a candidate gene responsible for the polydactylous mouse mutant Sasquatch. Current Biology, 9: 97-100.
Gould, A., et al (1998). Initiation of Rhombomeric Hoxb4 Expression Requires Induction by Somites and a Retinoic Pathway. Neuron, 21: 39-51.
Sharpe, J., et al (1998). Selectivity, sharing and competitive interactions in the regulation of Hoxb genes. EMBO, 17: 1788-1798.
Studer, M., et al (1998). Genetic interactions between Hoxa1 and Hoxb1 reveal new roles in regulation of early hindbrain patterning. Development, 125: 1025-1036.
Maconochie, M., et al (1997). Cross-regulation in the mouse HoxB complex: the expression of Hoxb-2 in rhombomere 4 is regulated by Hoxb-1. Genes and Development, 11: 1885-1895.
Aparicio, S., et al (1997). Organization of the Fugu rubripes Hox clusters, evidence for continuing evolution of vertebrate Hox complexes. Nature Genetics, 16: 79-84.
Manzanares, M., et al (1997). Segmental regulation of Hoxb-3 by kreisler. Nature, 387: 191-195.
Cohn, M., et al (1997). Hox9 genes and vertebrate limb specification. Nature, 387: 97-101.
Gould, A., et al (1997). Positive cross-regulation and enhancer sharing: two mechanisms for specifying overlapping Hox expression patterns. Genes and Development, 11: 900-913.
Studer,M., et al (1996). Altered segmental identity and abnormal migration of motor neurons in mice lacking Hoxb-1. Nature, 384: 630-634.
Vesque, C., et al (1996). Hoxb-2 transcriptional activation in rhombomeres 3 and 5 requires an evolutionarily conserved cis-acting element in addition to the Krox-20 binding site. EMBO J., 15: 5383-5396.
Itasaki, N., et al (1996). Reprogramming Hox Expression in the Vertebrate Hindbrain: Influence of Paraxial Mesoderm and Rhombomere Transposition. Neuron, 16: 487-500.
Nonchev, S. et al (1996). The conserved role of Krox-20 in directing Hox gene expression during vertebrate hindbrain segmentation. Proc. Natl. Acad. Sci. (USA), 93: 9339-9345.
Popperl, H., et al (995) Segmental Expression of Hoxb-1 Is Controlled by a Highly Conserved Autoregulatory Loop Dependent upon exd/Pbx. Cell. 81: 1031-1042.
Aparicio, S. et al (1995). Detecting conserved regulatory elements with the model genome of the Japanese puffer fish, Fugu rubripes. Proc. Natl. Acad. Sci. (USA) 92: 1684-1688.
Studer, M., et al (1994). Role of a Conserved Retinoic-Acid Response Element in Rhombomere Restriction of Hoxb-1. Science 265: 1728-1732.
Krumlauf, R. (1994). Hox Genes in Vertebrate Development. Cell 78: 191-201.
Marshall, H., et al (1994). A conserved retinoic acid response element required for early expression of the homeobox gene Hoxb-1. Nature 370: 567-571.
Ramirez-Solis, R., et al (1993). Hox-b4 (Hox-2.6) Mutant Mice Show Homeotic Transformation of Cervical Vertebra and Defects in the Closure of the Sternal Rudiments. Cell 73: 279-294.
Sham, M., et al (1993). The Zinc Finger Gene Krox-20 Regulates Hox-b2 (Hox-2.8) during Hindbrain Segmentation. Cell 72: 183-196.
Marshall, H., et al (1992). Retinoic acid alters hindbrain Hox code and induces transformation of rhombomeres 2/3 into a 4/5 identity. Nature 360: 737-741.
Sham, M.-H., et al (1992). Analysis of the murine Hox-2.7 gene: conserved alternative transcripts with differential distributions in the nervous system and the potential for shared regulatory regions. EMBO J. 11: 1825-1836.
Guthrie, S., et al (1992) Neuroectodermal autonomy of Hox-2.9 expression revealed by rhombomere transpositions. Nature 356: 157-159.
Whiting, J., et al (1991). Multiple spatially specific enhancers are required to reconstruct the pattern of Hox 2.6 gene expression. Genes and Development 5:2048-2059.
Hunt, P., et al (1991). A distinct Hox code for the branchial region of the vertebrate head. Nature. 353: 861-864.
Hasty, P., et al (1991). Introduction of a subtle mutation into the Hox 2.6 locus in embryonic stem cells. Nature 350: 243 246.
Graham, A., et al (1991). The murine Hox 2 genes display dynamic dorsoventral patterns of expression during central nervous system development. Development 112: 255 264.
Hunt, P., et al (1991). Patterning the vertebrate head: murine Hox 2 genes mark distinct subpopulations of premigratory and migrating cranial neural crest. Development 112: 43 50.
Hunt, P., and Krumlauf, R. (1991). Deciphering the Hox Code: Clues to Patterning the Branchial Region of the Head. Cell, 66: 1075-1078.
Wilkinson, D., et al (1989). Segmental expression of Hox 2 homeobox containing genes in the developing mouse hindbrain. Nature Vol. 341: 405 409.
Graham, A., Papalopulu, N., and Krumlauf, R. (1989). The Murine and Drosophila Homeobox Gene Complexes Have Common Features of Organization and Expression. Cell 57: 367 378.
Graham, A., et al (1988). Characterization of a murine homeo box gene, Hox 2.6, related to the Drosophila Deformed gene. Genes and Development. 2: 1424 1438.
Hammer, R., et al (1987) Diversity of Alpha Fetoprotein Gene Expression in Mice Is Generated by a Combination of Separate Enhancer Elements. Science 235: 53 58.
Krumlauf, R. et al (1986). Differential expression of a-fetoprotein genes on the inactive X chromosome in extraembryonic and somatic tissues of a transgenic mouse line. Nature 319: 224 226.