Network


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

Hotspot


Dive into the research topics where Elizabeth J. Thatcher is active.

Publication


Featured researches published by Elizabeth J. Thatcher.


Nature Genetics | 2007

Zebrafish miR-214 modulates Hedgehog signaling to specify muscle cell fate

Alex S. Flynt; Nan Li; Elizabeth J. Thatcher; Lilianna Solnica-Krezel; James G. Patton

Numerous microRNAs (miRNAs) have been discovered in the genomes of higher eukaryotes, and functional studies indicate that they are important during development. However, little is known concerning the function of individual miRNAs. We approached this problem in zebrafish by combining identification of miRNA expression, functional analyses and experimental validation of potential targets. We show that miR-214 is expressed during early segmentation stages in somites and that varying its expression alters the expression of genes regulated by Hedgehog signaling. Inhibition of miR-214 results in a reduction or loss of slow-muscle cell types. We show that su(fu) mRNA, encoding a negative regulator of Hedgehog signaling, is targeted by miR-214. Through regulation of su(fu), miR-214 enables precise specification of muscle cell types by sharpening cellular responses to Hedgehog.


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

Regulation of zebrafish fin regeneration by microRNAs.

Elizabeth J. Thatcher; Ima Paydar; Kimberly K. Anderson; James G. Patton

A number of genes have been implicated in regeneration, but the regulation of these genes, particularly pertaining to regeneration in higher vertebrates, remains an interesting and mostly open question. We have studied microRNA (miRNA) regulation of regeneration and found that an intact miRNA pathway is essential for caudal fin regeneration in zebrafish. We also showed that miR-203 directly targets the Wnt signaling transcription factor Lef1 during this process. Repression of Lef1 by miR-203 blocks regeneration, whereas loss of miR-203 results in excess Lef1 levels and fin overgrowth. Expression of Lef1 from mRNAs lacking 3′ UTR recognition elements can rescue the effects of excess miR-203, demonstrating that these effects are due to specific regulation of lef1 by miR-203. Our data support a model in which regulation of Lef1 protein levels by miR-203 is a key limiting step during regeneration.


Journal of Cell Biology | 2009

miR-8 microRNAs regulate the response to osmotic stress in zebrafish embryos

Alex S. Flynt; Elizabeth J. Thatcher; Kristopher Burkewitz; Nan Li; Yinzi Liu; James G. Patton

MicroRNAs (miRNAs) are highly conserved small RNAs that act as translational regulators of gene expression, exerting their influence by selectively targeting mRNAs bearing complementary sequence elements. These RNAs function in diverse aspects of animal development and physiology. Because of an ability to act as rapid responders at the level of translation, miRNAs may also influence stress response. In this study, we show that the miR-8 family of miRNAs regulates osmoregulation in zebrafish embryos. Ionocytes, which are a specialized cell type scattered throughout the epidermis, are responsible for pH and ion homeostasis during early development before gill formation. The highly conserved miR-8 family is expressed in ionocytes and enables precise control of ion transport by modulating the expression of Nherf1, which is a regulator of apical trafficking of transmembrane ion transporters. Ultimately, disruption of miR-8 family member function leads to an inability to respond to osmotic stress and blocks the ability to properly traffic and/or cluster transmembrane glycoproteins at the apical surface of ionocytes.


Developmental Dynamics | 2007

MiRNA expression analysis during normal zebrafish development and following inhibition of the Hedgehog and Notch signaling pathways.

Elizabeth J. Thatcher; Alex S. Flynt; Nan Li; Jonathan R. Patton; James G. Patton

microRNAs (miRNAs) are small (∼22 nucleotide) non‐coding RNAs that regulate gene expression at the post‐transcriptional level, typically by inhibiting translation. The genes encoding these small RNAs are estimated to comprise approximately 2–3% of animal genomes yet potentially regulate a majority of protein‐coding genes including those involved in cell specification and development. A key remaining question is to identify target mRNAs regulated by microRNAs. As a means to identify potential targets, we designed a sensitive microarray to analyze global miRNA expression patterns at twelve developmental stages in zebrafish. Further, we conducted arrays on zebrafish embryos treated with small molecule inhibitors of the Hedgehog and Notch signaling pathways to enable identification of differentially expressed miRNAs that target genes controlling key developmental pathways during early embryogenesis. Developmental Dynamics 236:2172–2180, 2007.


BMC Genomics | 2008

Genomic Organization of Zebrafish microRNAs

Elizabeth J. Thatcher; Jordan Bond; Ima Paydar; James G. Patton

BackgroundmicroRNAs (miRNAs) are small (~22 nt) non-coding RNAs that regulate cell movement, specification, and development. Expression of miRNAs is highly regulated, both spatially and temporally. Based on direct cloning, sequence conservation, and predicted secondary structures, a large number of miRNAs have been identified in higher eukaryotic genomes but whether these RNAs are simply a subset of a much larger number of noncoding RNA families is unknown. This is especially true in zebrafish where genome sequencing and annotation is not yet complete.ResultsWe analyzed the zebrafish genome to identify the number and location of proven and predicted miRNAs resulting in the identification of 35 new miRNAs. We then grouped all 415 zebrafish miRNAs into families based on seed sequence identity as a means to identify possible functional redundancy. Based on genomic location and expression analysis, we also identified those miRNAs that are likely to be encoded as part of polycistronic transcripts. Lastly, as a resource, we compiled existing zebrafish miRNA expression data and, where possible, listed all experimentally proven mRNA targets.ConclusionCurrent analysis indicates the zebrafish genome encodes 415 miRNAs which can be grouped into 44 families. The largest of these families (the miR-430 family) contains 72 members largely clustered in two main locations along chromosome 4. Thus far, most zebrafish miRNAs exhibit tissue specific patterns of expression.


PLOS ONE | 2013

miR-153 Regulates SNAP-25, Synaptic Transmission, and Neuronal Development

Chunyao Wei; Elizabeth J. Thatcher; Abigail F. Olena; Diana J. Cha; Ana Luisa Perdigoto; Andrew F. Marshall; Bruce D. Carter; Kendal Broadie; James G. Patton

SNAP-25 is a core component of the trimeric SNARE complex mediating vesicle exocytosis during membrane addition for neuronal growth, neuropeptide/growth factor secretion, and neurotransmitter release during synaptic transmission. Here, we report a novel microRNA mechanism of SNAP-25 regulation controlling motor neuron development, neurosecretion, synaptic activity, and movement in zebrafish. Loss of miR-153 causes overexpression of SNAP-25 and consequent hyperactive movement in early zebrafish embryos. Conversely, overexpression of miR-153 causes SNAP-25 down regulation resulting in near complete paralysis, mimicking the effects of treatment with Botulinum neurotoxin. miR-153-dependent changes in synaptic activity at the neuromuscular junction are consistent with the observed movement defects. Underlying the movement defects, perturbation of miR-153 function causes dramatic developmental changes in motor neuron patterning and branching. Together, our results indicate that precise control of SNAP-25 expression by miR-153 is critically important for proper neuronal patterning as well as neurotransmission.


RNA Biology | 2010

Small RNAs have a big impact on regeneration.

Elizabeth J. Thatcher; James G. Patton

A number of lower vertebrates including urodele amphibians and teleost fish are remarkably adept at repairing and regenerating damaged tissues and organs. Freshwater planarians are even more amazing, capable of regenerating entire body plans from small amputated fragments. In contrast, mammalian regenerative capacity is quite limited but of intense interest, especially related to human health and disease. For those organisms capable of robust regeneration, a common theme is the use of stem cells to replace complex tissues. Key questions remain as to the origin of these cells, whether there are pools of such cells that migrate to injured regions or whether they are generated on site. Beyond their origin, how are the genetic pathways that enable differentiation into multiple cell types and tissues regulated? microRNAs (miRNAs) are small noncoding RNAs that have recently been shown to play important roles in controlling stem cell self-renewal, proliferation, and differentiation. Some of these are thought to be required to maintain “stemness”. Here, we summarize recent work on the role of miRNAs in stem cells and their roles during regeneration.


Developmental Biology | 2015

miR-216a regulates snx5, a novel notch signaling pathway component, during zebrafish retinal development

Abigail F. Olena; Mahesh B. Rao; Elizabeth J. Thatcher; Shu-Yu Wu; James G. Patton

Precise regulation of Notch signaling is essential for normal vertebrate development. Mind bomb (Mib) is a ubiquitin ligase that is required for activation of Notch by Notch׳s ligand, Delta. Sorting Nexin 5 (SNX5) co-localizes with Mib and Delta complexes and has been shown to directly bind to Mib. We show that microRNA-216a (miR-216a) is expressed in the retina during early development and regulates snx5 to precisely regulate Notch signaling. miR-216a and snx5 have complementary expression patterns. Knocking down miR-216a and/or overexpression of snx5 resulted in increased Notch activation. Conversely, knocking down snx5 and/or miR-216a overexpression caused a decrease in Notch activation. We propose a model in which SNX5, precisely controlled by miR-216a, is a vital partner of Mib in promoting endocytosis of Delta and subsequent activation of Notch signaling.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2013

Strain-specific suppression of microRNA-320 by carcinogenic Helicobacter pylori promotes expression of the antiapoptotic protein Mcl-1

Jennifer M. Noto; M. Blanca Piazuelo; Rupesh Chaturvedi; Courtney Bartel; Elizabeth J. Thatcher; Alberto G. Delgado; Judith Romero-Gallo; Keith T. Wilson; Pelayo Correa; James G. Patton; Richard M. Peek

Helicobacter pylori is the strongest risk factor for gastric cancer, and strains harboring the cag pathogenicity island, which translocates the oncoprotein CagA into host cells, further augment cancer risk. We previously reported that in vivo adaptation of a noncarcinogenic H. pylori strain (B128) generated a derivative strain (7.13) with the ability to induce adenocarcinoma, providing a unique opportunity to define mechanisms that mediate gastric carcinogenesis. MicroRNAs (miRNAs) are small noncoding RNAs that regulate expression of oncogenes or tumor suppressors and are frequently dysregulated in carcinogenesis. To identify miRNAs and their targets involved in H. pylori-mediated carcinogenesis, miRNA microarrays were performed on RNA isolated from gastric epithelial cells cocultured with H. pylori strains B128, 7.13, or a 7.13 cagA(-) isogenic mutant. Among 61 miRNAs differentially expressed in a cagA-dependent manner, the tumor suppressor miR-320 was significantly downregulated by strain 7.13. Since miR-320 negatively regulates the antiapoptotic protein Mcl-1, we demonstrated that H. pylori significantly induced Mcl-1 expression in a cagA-dependent manner and that suppression of Mcl-1 results in increased apoptosis. To extend these results, mice were challenged with H. pylori strain 7.13 or its cagA(-) mutant; consistent with cell culture data, H. pylori induced Mcl-1 expression in a cagA-dependent manner. In human subjects, cag(+) strains induced significantly higher levels of Mcl-1 than cag(-) strains, and Mcl-1 expression levels paralleled the severity of neoplastic lesions. Collectively, these results indicate that H. pylori suppresses miR-320, upregulates Mcl-1, and decreases apoptosis in a cagA-dependent manner, which likely confers an increased risk for gastric carcinogenesis.


Archive | 2009

RNA Interference and microRNAs in Zebra Fish

Alex S. Flynt; Elizabeth J. Thatcher; James G. Patton

Collaboration


Dive into the Elizabeth J. Thatcher's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nan Li

Vanderbilt University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jennifer M. Noto

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Richard M. Peek

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Alberto G. Delgado

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge