Network


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

Hotspot


Dive into the research topics where Leticia Sansores-Garcia is active.

Publication


Featured researches published by Leticia Sansores-Garcia.


The EMBO Journal | 2011

Modulating F-actin organization induces organ growth by affecting the Hippo pathway.

Leticia Sansores-Garcia; Wouter Bossuyt; Ken Ichi Wada; Shigenobu Yonemura; Chunyao Tao; Hiroshi Sasaki; Georg Halder

The Hippo tumour suppressor pathway is a conserved signalling pathway that controls organ size. The core of the Hpo pathway is a kinase cascade, which in Drosophila involves the Hpo and Warts kinases that negatively regulate the activity of the transcriptional coactivator Yorkie. Although several additional components of the Hippo pathway have been discovered, the inputs that regulate Hippo signalling are not fully understood. Here, we report that induction of extra F‐actin formation, by loss of Capping proteins A or B, or caused by overexpression of an activated version of the formin Diaphanous, induced strong overgrowth in Drosophila imaginal discs through modulating the activity of the Hippo pathway. Importantly, loss of Capping proteins and Diaphanous overexpression did not significantly affect cell polarity and other signalling pathways, including Hedgehog and Decapentaplegic signalling. The interaction between F‐actin and Hpo signalling is evolutionarily conserved, as the activity of the mammalian Yorkie‐orthologue Yap is modulated by changes in F‐actin. Thus, regulators of F‐actin, and in particular Capping proteins, are essential for proper growth control by affecting Hippo signalling.


Proceedings of the National Academy of Sciences of the United States of America | 2010

The apical-basal cell polarity determinant Crumbs regulates Hippo signaling in Drosophila

Chiao Lin Chen; Kathleen Gajewski; Fisun Hamaratoglu; Wouter Bossuyt; Leticia Sansores-Garcia; Chunyao Tao; Georg Halder

Defects in apical-basal cell polarity and abnormal expression of cell polarity determinants are often associated with cancer in vertebrates. In Drosophila, abnormal expression of apical-basal determinants can cause neoplastic phenotypes, including loss of cell polarity and overproliferation. However, the pathways through which apical-basal polarity determinants affect growth are poorly understood. Here, we investigated the mechanism by which the apical determinant Crumbs (Crb) affects growth in Drosophila imaginal discs. Overexpression of Crb causes severe overproliferation, and we found that loss of Crb similarly results in overgrowth of imaginal discs. Crb gain and loss of function caused defects in Hippo signaling, a key signaling pathway that controls tissue growth in Drosophila and mammals. Manipulation of Crb levels caused the up-regulation of Hippo target genes, genetically interacted with known Hippo pathway components, and required Yorkie, a transcriptional coactivator that acts downstream in the Hippo pathway, for target gene induction and overgrowth. Interestingly, Crb regulates growth and cell polarity through different motifs in its intracellular domain. A juxtamembrane FERM domain-binding motif is responsible for growth regulation and induction of Hippo target gene expression, whereas Crb uses a PDZ-binding motif to form a complex with other polarity factors. The Hippo pathway component Expanded, an apically localized adaptor protein, is mislocalized in both crb mutant cells and Crb overexpressing tissues, whereas the other Hippo pathway components, Fat and Merlin, are unaffected. Taken together, our data show that Crb regulates growth through Hippo signaling, and thus identify Crb as a previously undescribed upstream input into the Hippo pathway.


Journal of Biological Chemistry | 2007

The Mast Cell-restricted Tryptase mMCP-6 Has a Critical Immunoprotective Role in Bacterial Infections

Shakeel M. Thakurdas; Ernestina Melicoff; Leticia Sansores-Garcia; Daniel C. Moreira; Youlia Petrova; Richard L. Stevens; Roberto Adachi

Although it has been shown that mast cell-deficient mice have diminished innate immune responses against bacteria, the most important immunoprotective factors secreted from activated mast cells have not been identified. Mouse mast cell protease 6 is a tetramer-forming tryptase. This serine protease is abundant in the secretory granules and is exocytosed upon bacterial challenge. Here we have described the generation of a mast cell protease-6-null mouse. Our discovery that mice lacking this neutral protease cannot efficiently clear Klebsiella pneumoniae from their peritoneal cavities reveals an essential role for this serine protease, and presumably its human ortholog, in innate immunity.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Boundaries of Dachsous Cadherin activity modulate the Hippo signaling pathway to induce cell proliferation

Maria Willecke; Fisun Hamaratoglu; Leticia Sansores-Garcia; Chunyao Tao; Georg Halder

The conserved Hippo tumor suppressor pathway is a key signaling pathway that controls organ size in Drosophila. To date a signal transduction cascade from the Cadherin Fat at the plasma membrane into the nucleus has been discovered. However, how the Hippo pathway is regulated by extracellular signals is poorly understood. Fat not only regulates growth but also planar cell polarity, for which it interacts with the Dachsous (Ds) Cadherin, and Four-jointed (Fj), a transmembrane kinase that modulates the interaction between Ds and Fat. Ds and Fj are expressed in gradients and manipulation of their expression causes abnormal growth. However, how Ds and Fj regulate growth and whether they act through the Hippo pathway is not known. Here, we report that Ds and Fj regulate Hippo signaling to control growth. Interestingly, we found that Ds/Fj regulate the Hippo pathway through a remarkable logic. Induction of Hippo target genes is not proportional to the amount of Ds or Fj presented to a cell, as would be expected if Ds and Fj acted as traditional ligands. Rather, Hippo target genes are up-regulated when neighboring cells express different amounts of Ds or Fj. Consistent with a model that differences in Ds/Fj levels between cells regulate the Hippo pathway, we found that artificial Ds/Fj boundaries induce extra cell proliferation, whereas flattening the endogenous Ds and Fj gradients results in growth defects. The Ds/Fj signaling system thus defines a cell-to-cell signaling mechanism that regulates the Hippo pathway, thereby contributing to the control of organ size.


Journal of Cell Science | 2009

The Hippo tumor-suppressor pathway regulates apical-domain size in parallel to tissue growth.

Fisun Hamaratoglu; Kathleen Gajewski; Leticia Sansores-Garcia; Clayton M. Morrison; Chunyao Tao; Georg Halder

The Hippo tumor-suppressor pathway controls tissue growth in Drosophila and mammals by regulating cell proliferation and apoptosis. The Hippo pathway includes the Fat cadherin, a transmembrane protein, which acts upstream of several other components that form a kinase cascade that culminates in the regulation of gene expression through the transcriptional coactivator Yorkie (Yki). Our previous work in Drosophila indicated that Merlin (Mer) and Expanded (Ex) are members of the Hippo pathway and act upstream of the Hippo kinase. In contrast to this model, it was suggested that Mer and Ex primarily regulate membrane dynamics and receptor trafficking, thereby affecting Hippo pathway activity only indirectly. Here, we examined the effects of Mer, Ex and the Hippo pathway on the size of the apical membrane and on apical-basal polarity complexes. We found that mer;ex double mutant imaginal disc cells have significantly increased levels of apical membrane determinants, such as Crb, aPKC and Patj. These phenotypes were shared with mutations in other Hippo pathway components and required Yki, indicating that Mer and Ex signal through the Hippo pathway. Interestingly, however, whereas Crb was required for the accumulation of other apical proteins and for the expansion of the apical domain observed in Hippo pathway mutants, its elimination did not significantly reverse the overgrowth phenotype of warts mutant cells. Therefore, Hippo signaling regulates cell polarity complexes in addition to and independently of its growth control function in imaginal disc cells.


Journal of Biological Chemistry | 2009

Synaptotagmin-2 Controls Regulated Exocytosis but Not Other Secretory Responses of Mast Cells

Ernestina Melicoff; Leticia Sansores-Garcia; Alejandra Gomez; Daniel C. Moreira; Proleta Datta; Pratima Thakur; Youlia Petrova; Tanya Siddiqi; Jayasimha N. Murthy; Burton F. Dickey; Ruth Heidelberger; Roberto Adachi

Mast cell degranulation is a highly regulated, calcium-dependent process, which is important for the acute release of inflammatory mediators during the course of many pathological conditions. We previously found that Synaptotagmin-2, a calcium sensor in neuronal exocytosis, was expressed in a mast cell line. We postulated that this protein may be involved in the control of mast cell-regulated exocytosis, and we generated Synaptotagmin-2 knock-out mice to test our hypothesis. Mast cells from this mutant animal conferred an abnormally decreased passive cutaneous anaphylaxis reaction on mast cell-deficient mice that correlated with a specific defect in mast cell-regulated exocytosis, leaving constitutive exocytosis and nonexocytic mast cell effector responses intact. This defect was not secondary to abnormalities in the development, maturation, migration, morphology, synthesis, and storage of inflammatory mediators, or intracellular calcium transients of the mast cells. Unlike neurons, the lack of Synaptotagmin-2 in mast cells was not associated with increased spontaneous exocytosis.


Current Biology | 2013

Mask Is Required for the Activity of the Hippo Pathway Effector Yki/YAP

Leticia Sansores-Garcia; Mardelle Atkins; Iván M. Moya; Maria Shahmoradgoli; Chunyao Tao; Gordon B. Mills; Georg Halder

The Drosophila Yorkie (Yki) protein and its mammalian homolog Yes-associated protein (YAP) are potent growth promoters, and YAP overexpression is associated with multiple types of cancer. Yki and YAP are transcriptional coactivators and function as downstream effectors of the Hippo tumor suppressor pathway. The regulation of Yki and YAP by the Hippo signaling pathway has been extensively investigated; however, how they regulate gene expression is poorly understood. To identify additional regulators of Yki activity, we performed a genome-wide RNAi screen in Drosophila S2 cells. In this screen, we identified the conserved protein Mask (Multiple ankyrin repeats single KH domain) as a novel promoter of Yki activity in vitro and validated this function in vivo in Drosophila. We found that Mask is required downstream of the Hippo pathway for Yki to induce target-gene expression and that Mask forms complexes with Yki. The human Mask homolog MASK1 complexes with YAP and is required for the full activity of YAP. Additionally, elevated MASK1 expression is associated with worsened outcomes for breast cancer patients. We conclude that Mask is a novel cofactor for Yki/YAP required for optimal Yki/YAP activity during development and oncogenesis.


PLOS Genetics | 2013

Dynamic Rewiring of the Drosophila Retinal Determination Network Switches Its Function from Selector to Differentiation

Mardelle Atkins; Yuwei Jiang; Leticia Sansores-Garcia; Barbara Jusiak; Georg Halder; Graeme Mardon

Organ development is directed by selector gene networks. Eye development in the fruit fly Drosophila melanogaster is driven by the highly conserved selector gene network referred to as the “retinal determination gene network,” composed of approximately 20 factors, whose core comprises twin of eyeless (toy), eyeless (ey), sine oculis (so), dachshund (dac), and eyes absent (eya). These genes encode transcriptional regulators that are each necessary for normal eye development, and sufficient to direct ectopic eye development when misexpressed. While it is well documented that the downstream genes so, eya, and dac are necessary not only during early growth and determination stages but also during the differentiation phase of retinal development, it remains unknown how the retinal determination gene network terminates its functions in determination and begins to promote differentiation. Here, we identify a switch in the regulation of ey by the downstream retinal determination genes, which is essential for the transition from determination to differentiation. We found that central to the transition is a switch from positive regulation of ey transcription to negative regulation and that both types of regulation require so. Our results suggest a model in which the retinal determination gene network is rewired to end the growth and determination stage of eye development and trigger terminal differentiation. We conclude that changes in the regulatory relationships among members of the retinal determination gene network are a driving force for key transitions in retinal development.


Proceedings of the National Academy of Sciences of the United States of America | 2018

Modulation of the Hippo pathway and organ growth by RNA processing proteins

Jana Mach; Mardelle Atkins; Kathleen Gajewski; Violaine Mottier-Pavie; Leticia Sansores-Garcia; Jun Xie; Robert Andrew Mills; Weronika Kowalczyk; Leen Van Huffel; Gordon B. Mills; Georg Halder

Significance The Hippo pathway is an important regulator of animal growth, and its effector, Yorkie (in flies) or YAP/TAZ (in mammals), drives the expression of genes needed for cell proliferation and survival. In an effort to identify new regulators of Yorkie, we performed a genetic modifier screen in Drosophila. In this screen, we identified the RNA-binding protein Hrb27C as a positive regulator of Yorkie activity that modulates its phosphorylation status. Additional experiments identified the Hrb27C interacting proteins Glorund, Halfpint, Squid, and Pabp2 as negative modulators of Yorkie activity. Our results identify a link between the Hippo pathway and RNA binding factors and deepen the knowledge of this important growth control pathway. The Hippo tumor-suppressor pathway regulates organ growth, cell proliferation, and stem cell biology. Defects in Hippo signaling and hyperactivation of its downstream effectors—Yorkie (Yki) in Drosophila and YAP/TAZ in mammals—result in progenitor cell expansion and overgrowth of multiple organs and contribute to cancer development. Deciphering the mechanisms that regulate the activity of the Hippo pathway is key to understanding its function and for therapeutic targeting. However, although the Hippo kinase cascade and several other upstream inputs have been identified, the mechanisms that regulate Yki/YAP/TAZ activity are still incompletely understood. To identify new regulators of Yki activity, we screened in Drosophila for suppressors of tissue overgrowth and Yki activation caused by overexpression of atypical protein kinase C (aPKC), a member of the apical cell polarity complex. In this screen, we identified mutations in the heterogeneous nuclear ribonucleoprotein Hrb27C that strongly suppressed the tissue defects induced by ectopic expression of aPKC. Hrb27C was required for aPKC-induced tissue growth and Yki target gene expression but did not affect general gene expression. Genetic and biochemical experiments showed that Hrb27C affects Yki phosphorylation. Other RNA-binding proteins known to interact with Hrb27C for mRNA transport in oocytes were also required for normal Yki activity, although they suppressed Yki output. Based on the known functions of Hrb27C, we conclude that Hrb27C-mediated control of mRNA splicing, localization, or translation is essential for coordinated activity of the Hippo pathway.


Developmental Cell | 2017

Hippo Reprograms the Transcriptional Response to Ras Signaling

Justine Pascual; Jelle Jacobs; Leticia Sansores-Garcia; Malini Natarajan; Julia Zeitlinger; Stein Aerts; Georg Halder; Fisun Hamaratoglu

Collaboration


Dive into the Leticia Sansores-Garcia's collaboration.

Top Co-Authors

Avatar

Georg Halder

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Chunyao Tao

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kathleen Gajewski

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ernestina Melicoff

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Mardelle Atkins

Sam Houston State University

View shared research outputs
Top Co-Authors

Avatar

Roberto Adachi

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel C. Moreira

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Gordon B. Mills

Sam Houston State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge