Network


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

Hotspot


Dive into the research topics where Oleg Lagutin is active.

Publication


Featured researches published by Oleg Lagutin.


The EMBO Journal | 2006

Six2 is required for suppression of nephrogenesis and progenitor renewal in the developing kidney

Michelle Self; Oleg Lagutin; Beth Bowling; Jaime Hendrix; Yi Cai; Gregory R. Dressler; Guillermo Oliver

During kidney development and in response to inductive signals, the metanephric mesenchyme aggregates, becomes polarized, and generates much of the epithelia of the nephron. As such, the metanephric mesenchyme is a renal progenitor cell population that must be replenished as epithelial derivatives are continuously generated. The molecular mechanisms that maintain the undifferentiated state of the metanephric mesenchymal precursor cells have not yet been identified. In this paper, we report that functional inactivation of the homeobox gene Six2 results in premature and ectopic differentiation of mesenchymal cells into epithelia and depletion of the progenitor cell population within the metanephric mesenchyme. Failure to renew the mesenchymal cells results in severe renal hypoplasia. Gain of Six2 function in cortical metanephric mesenchymal cells was sufficient to prevent their epithelial differentiation in an organ culture assay. We propose that in the developing kidney, Six2 activity is required for maintaining the mesenchymal progenitor population in an undifferentiated state by opposing the inductive signals emanating from the ureteric bud.


Genes & Development | 2010

The nuclear hormone receptor Coup-TFII is required for the initiation and early maintenance of Prox1 expression in lymphatic endothelial cells

R. Sathish Srinivasan; Xin Geng; Ying Yang; Yingdi Wang; Suraj Mukatira; Michèle Studer; Marianna P.R. Porto; Oleg Lagutin; Guillermo Oliver

The homeobox gene Prox1 is crucial for mammalian lymphatic vascular development. In the absence of Prox1, lymphatic endothelial cells (LECs) are not specified. The maintenance of LEC identity also requires the constant expression of Prox1. However, the mechanisms controlling the expression of this gene in LECs remain poorly understood. The SRY-related gene Sox18 is required to induce Prox1 expression in venous LEC progenitors. Although Sox18 is also expressed in embryonic arteries, these vessels do not express Prox1, nor do they give rise to LECs. This finding suggests that some venous endothelial cell-specific factor is required for the activation of Prox1. Here we demonstrate that the nuclear hormone receptor Coup-TFII is necessary for the activation of Prox1 in embryonic veins by directly binding a conserved DNA domain in the regulatory region of Prox1. In addition, we show that the direct interaction between nuclear hormone receptors and Prox1 is also necessary for the maintenance of Prox1 expression during early stages of LEC specification and differentiation.


PLOS Biology | 2010

Prox1 is required for granule cell maturation and intermediate progenitor maintenance during brain neurogenesis.

Alfonso Lavado; Oleg Lagutin; Lionel M.L. Chow; Suzanne J. Baker; Guillermo Oliver

The transcription factor Prox1 plays a crucial role in intermediate progenitor maintenance and hippocampal neuron differentiation during adult neurogenesis in the dentate gyrus region of the hippocampus.


Developmental Cell | 2008

Haploinsufficiency of Six3 Fails to Activate Sonic hedgehog Expression in the Ventral Forebrain and Causes Holoprosencephaly

Xin Geng; Christina K. Speirs; Oleg Lagutin; Wei Liu; Lilianna Solnica-Krezel; Yongsu Jeong; Douglas J. Epstein; Guillermo Oliver

Holoprosencephaly (HPE), the most common forebrain malformation, is characterized by an incomplete separation of the cerebral hemispheres. Mutations in the homeobox gene SIX3 account for 1.3% of all cases of human HPE. Using zebrafish-based assays, we have now determined that HPE-associated Six3 mutant proteins function as hypomorphs. Haploinsufficiency of Six3 caused by deletion of one allele of Six3 or by replacement of wild-type Six3 with HPE-associated Six3 mutant alleles was sufficient to recapitulate in mouse models most of the phenotypic features of human HPE. We demonstrate that Shh is a direct target of Six3 in the rostral diencephalon ventral midline (RDVM). Reduced amounts of functional Six3 protein fail to activate Shh expression in the mutant RDVM and ultimately lead to HPE. These results identify Six3 as a direct regulator of Shh expression and reveal a crossregulatory loop between Shh and Six3 in the ventral forebrain.


The EMBO Journal | 2006

Six3 activation of Pax6 expression is essential for mammalian lens induction and specification

Wei Liu; Oleg Lagutin; Michael Mende; Andrea Streit; Guillermo Oliver

The homeobox gene Six3 regulates forebrain development. Here we show that Six3 is also crucial for lens formation. Conditional deletion of mouse Six3 in the presumptive lens ectoderm (PLE) disrupted lens formation. In the most severe cases, lens induction and specification were defective, and the lens placode and lens were absent. In Six3‐mutant embryos, Pax6 was downregulated, and Sox2 was absent in the lens preplacodal ectoderm. Using ChIP, electrophoretic mobility shift assay, and luciferase reporter assays, we determined that Six3 activates Pax6 and Sox2 expression. Misexpression of mouse Six3 into chick embryos promoted the ectopic expansion of the ectodermal Pax6 expression domain. Our results position Six3 at the top of the regulatory pathway leading to lens formation. We conclude that Six3 directly activates Pax6 and probably also Sox2 in the PLE and regulates cell autonomously the earliest stages of mammalian lens induction.


Developmental Dynamics | 2001

Six3 promotes the formation of ectopic optic vesicle-like structures in mouse embryos.

Oleg Lagutin; Changqi C. Zhu; Yasuhide Furuta; David H. Rowitch; Andrew P. McMahon; Guillermo Oliver

A few years ago, three novel murine homeobox genes closely related to the Drosophila sine oculis (so) gene (Six1‐3) were isolated and were all included in the Six/so gene family. Because of its early expression in the developing eye field, Six3 was initially thought to be the functional ortholog of the Drosophila so gene. This hypothesis was further supported by the demonstration that ectopic Six3 expression in medaka fish (Oryzias latipes) promotes the formation of ectopic lens and retina tissue. Here, we show that similar to Drosophila, where the eyeless/Pax6 gene regulates the eye‐specific expression of so, Six3 expression in the murine lens placodal ectoderm is also controlled by Pax6. We also show that ectopic Six3 expression promotes the formation of ectopic optic vesicle‐like structures in the hindbrain‐midbrain region of developing mouse embryos.


Development | 2008

Six3 inactivation causes progressive caudalization and aberrant patterning of the mammalian diencephalon

Alfonso Lavado; Oleg Lagutin; Guillermo Oliver

The homeobox gene Six3 represses Wnt1 transcription. It is also required in the anterior neural plate for the development of the mammalian rostral forebrain. We have now determined that at the 15- to 17-somite stage, the prospective diencephalon is the most-anterior structure in the Six3-null brain, and Wnt1 expression is anteriorly expanded. Consequently, the brain caudalizes, and at the 22- to 24-somite stage, the prospective thalamic territory is the most-anterior structure. At around E11.0, the pretectum replaces this structure. Analysis of Six3;Wnt1 double-null mice revealed that Six3-mediated repression of Wnt1 is necessary for the formation of the rostral diencephalon and that Six3 activity is required for the formation of the telencephalon. These results provide insight into the mechanisms that establish anteroposterior identity in the developing mammalian brain.


Journal of Clinical Investigation | 2010

Neuroretina specification in mouse embryos requires Six3-mediated suppression of Wnt8b in the anterior neural plate

Wei Liu; Oleg Lagutin; Eric C. Swindell; Milan Jamrich; Guillermo Oliver

Retinal degeneration causes vision impairment and blindness in humans. If one day we are to harness the potential of stem cell-based cell replacement therapies to treat these conditions, it is imperative that we better understand normal retina development. Currently, the genes and mechanisms that regulate the specification of the neuroretina during vertebrate eye development remain unknown. Here, we identify sine oculis-related homeobox 3 (Six3) as a crucial player in this process in mice. In Six3 conditional-mutant mouse embryos, specification of the neuroretina was abrogated, but that of the retinal pigmented epithelium was normal. Conditional deletion of Six3 did not affect the initial development of the optic vesicle but did arrest subsequent neuroretina specification. Ectopic rostral expansion of Wnt8b expression was the major response to Six3 deletion and the leading cause for the specific lack of neuroretina, as ectopic Wnt8b expression in transgenic embryos was sufficient to suppress neuroretina specification. Using chromatin immunoprecipitation assays, we identified Six3-responsive elements in the Wnt8b locus and demonstrated that Six3 directly repressed Wnt8b expression in vivo. Our findings provide a molecular framework to the program leading to neuroretina differentiation and may be relevant for the development of novel strategies aimed at characterizing and eventually treating different abnormalities in eye formation.


Development | 2016

Six3 dosage mediates the pathogenesis of holoprosencephaly.

Xin Geng; Sandra Acosta; Oleg Lagutin; Hyea Jin Gil; Guillermo Oliver

Holoprosencephaly (HPE) is defined as the incomplete separation of the two cerebral hemispheres. The pathology of HPE is variable and, based on the severity of the defect, HPE is divided into alobar, semilobar, and lobar. Using a novel hypomorphic Six3 allele, we demonstrate in mice that variability in Six3 dosage results in different HPE phenotypes. Furthermore, we show that whereas the semilobar phenotype results from severe downregulation of Shh expression in the rostral diencephalon ventral midline, the alobar phenotype is caused by downregulation of Foxg1 expression in the anterior neural ectoderm. Consistent with these results, in vivo activation of the Shh signaling pathway rescued the semilobar phenotype but not the alobar phenotype. Our findings show that variations in Six3 dosage result in different forms of HPE. Summary: Analysis of a hypomorphic Six3 allele reveals that variability in Six3 dosage is sufficient to generate mice exhibiting a spectrum of holoprosencephaly forms in a non-strain-specific manner.


Genes & Development | 2003

Six3 repression of Wnt signaling in the anterior neuroectoderm is essential for vertebrate forebrain development

Oleg Lagutin; Changqi C. Zhu; Daisuke Kobayashi; Jacek Topczewski; Kenji Shimamura; Luis Puelles; H. R. Russell; Peter J. McKinnon; Lilianna Solnica-Krezel; Guillermo Oliver

Collaboration


Dive into the Oleg Lagutin's collaboration.

Top Co-Authors

Avatar

Guillermo Oliver

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Wei Liu

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Xin Geng

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Alfonso Lavado

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lilianna Solnica-Krezel

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Yasuhide Furuta

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

R. Sathish Srinivasan

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Yingdi Wang

St. Jude Children's Research Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge