Network


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

Hotspot


Dive into the research topics where Walter L. Eckalbar is active.

Publication


Featured researches published by Walter L. Eckalbar.


PLOS ONE | 2014

Transcriptomic Analysis of Tail Regeneration in the Lizard Anolis carolinensis Reveals Activation of Conserved Vertebrate Developmental and Repair Mechanisms

Elizabeth D. Hutchins; Glenn J. Markov; Walter L. Eckalbar; Rajani M. George; Jesse M. King; Minami A. Tokuyama; Lauren A. Geiger; Nataliya Emmert; Michael J. Ammar; April N. Allen; Ashley L. Siniard; Jason J. Corneveaux; Rebecca E. Fisher; Juli Wade; Dale F. DeNardo; J. Alan Rawls; Matthew J. Huentelman; Jeanne Wilson-Rawls; Kenro Kusumi

Lizards, which are amniote vertebrates like humans, are able to lose and regenerate a functional tail. Understanding the molecular basis of this process would advance regenerative approaches in amniotes, including humans. We have carried out the first transcriptomic analysis of tail regeneration in a lizard, the green anole Anolis carolinensis, which revealed 326 differentially expressed genes activating multiple developmental and repair mechanisms. Specifically, genes involved in wound response, hormonal regulation, musculoskeletal development, and the Wnt and MAPK/FGF pathways were differentially expressed along the regenerating tail axis. Furthermore, we identified 2 microRNA precursor families, 22 unclassified non-coding RNAs, and 3 novel protein-coding genes significantly enriched in the regenerating tail. However, high levels of progenitor/stem cell markers were not observed in any region of the regenerating tail. Furthermore, we observed multiple tissue-type specific clusters of proliferating cells along the regenerating tail, not localized to the tail tip. These findings predict a different mechanism of regeneration in the lizard than the blastema model described in the salamander and the zebrafish, which are anamniote vertebrates. Thus, lizard tail regrowth involves the activation of conserved developmental and wound response pathways, which are potential targets for regenerative medical therapies.


BMC Genomics | 2013

Genome reannotation of the lizard Anolis carolinensis based on 14 adult and embryonic deep transcriptomes

Walter L. Eckalbar; Elizabeth D. Hutchins; Glenn J. Markov; April N. Allen; Jason J. Corneveaux; Kerstin Lindblad-Toh; Federica Di Palma; Jessica Alföldi; Matthew J. Huentelman; Kenro Kusumi

BackgroundThe green anole lizard, Anolis carolinensis, is a key species for both laboratory and field-based studies of evolutionary genetics, development, neurobiology, physiology, behavior, and ecology. As the first non-avian reptilian genome sequenced, A. carolinesis is also a prime reptilian model for comparison with other vertebrate genomes. The public databases of Ensembl and NCBI have provided a first generation gene annotation of the anole genome that relies primarily on sequence conservation with related species. A second generation annotation based on tissue-specific transcriptomes would provide a valuable resource for molecular studies.ResultsHere we provide an annotation of the A. carolinensis genome based on de novo assembly of deep transcriptomes of 14 adult and embryonic tissues. This revised annotation describes 59,373 transcripts, compared to 16,533 and 18,939 currently for Ensembl and NCBI, and 22,962 predicted protein-coding genes. A key improvement in this revised annotation is coverage of untranslated region (UTR) sequences, with 79% and 59% of transcripts containing 5’ and 3’ UTRs, respectively. Gaps in genome sequence from the current A. carolinensis build (Anocar2.0) are highlighted by our identification of 16,542 unmapped transcripts, representing 6,695 orthologues, with less than 70% genomic coverage.ConclusionsIncorporation of tissue-specific transcriptome sequence into the A. carolinensis genome annotation has markedly improved its utility for comparative and functional studies. Increased UTR coverage allows for more accurate predicted protein sequence and regulatory analysis. This revised annotation also provides an atlas of gene expression specific to adult and embryonic tissues.


Nature Communications | 2015

A PAX1 enhancer locus is associated with susceptibility to idiopathic scoliosis in females

Swarkar Sharma; Douglas Londono; Walter L. Eckalbar; Xiaochong Gao; Dongping Zhang; Kristen N. Mauldin; Ikuyo Kou; Atsushi Takahashi; Morio Matsumoto; Nobuhiro Kamiya; Karl K. Murphy; Reuel Cornelia; John A. Herring; Dennis K. Burns; Nadav Ahituv; Shiro Ikegawa; Derek Gordon; Carol A. Wise

Idiopathic scoliosis (IS) is a common paediatric musculoskeletal disease that displays a strong female bias. By performing a genome-wide association study (GWAS) of 3,102 individuals, we identify significant associations with 20p11.22 SNPs for females (P=6.89 × 10−9) but not males (P=0.71). This association with IS is also found in independent female cohorts from the United States of America and Japan (overall P=2.15 × 10−10, OR=1.30 (rs6137473)). Unexpectedly, the 20p11.22 IS risk alleles were previously associated with protection from early-onset alopecia, another sexually dimorphic condition. The 174-kb associated locus is distal to PAX1, which encodes paired box 1, a transcription factor involved in spine development. We identify a sequence in the associated locus with enhancer activity in zebrafish somitic muscle and spinal cord, an activity that is abolished by IS-associated SNPs. We thus identify a sexually dimorphic IS susceptibility locus, and propose the first functionally defined candidate mutations in an enhancer that may regulate expression in specific spinal cells.


Nature Communications | 2015

A genome-wide association study identifies four novel susceptibility loci underlying inguinal hernia

Eric Jorgenson; Nadja Makki; Ling Shen; David C. Chen; Chao Tian; Walter L. Eckalbar; David A. Hinds; Nadav Ahituv; Andrew Avins

Inguinal hernia repair is one of the most commonly performed operations in the world, yet little is known about the genetic mechanisms that predispose individuals to develop inguinal hernias. We perform a genome-wide association analysis of surgically confirmed inguinal hernias in 72,805 subjects (5,295 cases and 67,510 controls) and confirm top associations in an independent cohort of 92,444 subjects with self-reported hernia repair surgeries (9,701 cases and 82,743 controls). We identify four novel inguinal hernia susceptibility loci in the regions of EFEMP1, WT1, EBF2 and ADAMTS6. Moreover, we observe expression of all four genes in mouse connective tissue and network analyses show an important role for two of these genes (EFEMP1 and WT1) in connective tissue maintenance/homoeostasis. Our findings provide insight into the aetiology of hernia development and highlight genetic pathways for studies of hernia development and its treatment.


Nature Genetics | 2016

Transcriptomic and epigenomic characterization of the developing bat wing

Walter L. Eckalbar; Stephen A. Schlebusch; Mandy K. Mason; Zoe Gill; Ash V Parker; Betty M. Booker; Sierra Nishizaki; Christiane Muswamba-Nday; Elizabeth Terhune; Kimberly A. Nevonen; Nadja Makki; Tara Friedrich; Julia E. VanderMeer; Katherine S. Pollard; Lucia Carbone; Jeffrey D. Wall; Nicola Illing; Nadav Ahituv

Bats are the only mammals capable of powered flight, but little is known about the genetic determinants that shape their wings. Here we generated a genome for Miniopterus natalensis and performed RNA-seq and ChIP-seq (H3K27ac and H3K27me3) analyses on its developing forelimb and hindlimb autopods at sequential embryonic stages to decipher the molecular events that underlie bat wing development. Over 7,000 genes and several long noncoding RNAs, including Tbx5-as1 and Hottip, were differentially expressed between forelimb and hindlimb, and across different stages. ChIP-seq analysis identified thousands of regions that are differentially modified in forelimb and hindlimb. Comparative genomics found 2,796 bat-accelerated regions within H3K27ac peaks, several of which cluster near limb-associated genes. Pathway analyses highlighted multiple ribosomal proteins and known limb patterning signaling pathways as differentially regulated and implicated increased forelimb mesenchymal condensation in differential growth. In combination, our work outlines multiple genetic components that likely contribute to bat wing formation, providing insights into this morphological innovation.


Wiley Interdisciplinary Reviews-Developmental Biology | 2012

Scoliosis and segmentation defects of the vertebrae.

Walter L. Eckalbar; Rebecca E. Fisher; Alan Rawls; Kenro Kusumi

The vertebral column derives from somites, which are transient paired segments of mesoderm that surround the neural tube in the early embryo. Somites are formed by a genetic mechanism that is regulated by cyclical expression of genes in the Notch, Wnt, and fibroblast growth factor (FGF) signaling pathways. These oscillators together with signaling gradients within the presomitic mesoderm help to set somitic boundaries and rostral–caudal polarity that are essential for the precise patterning of the vertebral column. Disruption of this mechanism has been identified as the cause of severe segmentation defects of the vertebrae in humans. These segmentation defects are part of a spectrum of spinal disorders affecting the skeletal elements and musculature of the spine, resulting in curvatures such as scoliosis, kyphosis, and lordosis. While the etiology of most disorders with spinal curvatures is still unknown, genetic and developmental studies of somitogenesis and patterning of the axial skeleton and musculature are yielding insights into the causes of these diseases. WIREs Dev Biol 2012, 1:401–423. doi: 10.1002/wdev.34


PLOS Genetics | 2014

Genome-Wide Discovery of Drug-Dependent Human Liver Regulatory Elements

Robin P. Smith; Walter L. Eckalbar; Kari M. Morrissey; Marcelo R. Luizon; Thomas J. Hoffmann; Xuefeng Sun; Stacy L. Jones; Shelley Force Aldred; Anuradha Ramamoorthy; Zeruesenay Desta; Yunlong Liu; Todd C. Skaar; Nathan D. Trinklein; Kathleen M. Giacomini; Nadav Ahituv

Inter-individual variation in gene regulatory elements is hypothesized to play a causative role in adverse drug reactions and reduced drug activity. However, relatively little is known about the location and function of drug-dependent elements. To uncover drug-associated elements in a genome-wide manner, we performed RNA-seq and ChIP-seq using antibodies against the pregnane X receptor (PXR) and three active regulatory marks (p300, H3K4me1, H3K27ac) on primary human hepatocytes treated with rifampin or vehicle control. Rifampin and PXR were chosen since they are part of the CYP3A4 pathway, which is known to account for the metabolism of more than 50% of all prescribed drugs. We selected 227 proximal promoters for genes with rifampin-dependent expression or nearby PXR/p300 occupancy sites and assayed their ability to induce luciferase in rifampin-treated HepG2 cells, finding only 10 (4.4%) that exhibited drug-dependent activity. As this result suggested a role for distal enhancer modules, we searched more broadly to identify 1,297 genomic regions bearing a conditional PXR occupancy as well as all three active regulatory marks. These regions are enriched near genes that function in the metabolism of xenobiotics, specifically members of the cytochrome P450 family. We performed enhancer assays in rifampin-treated HepG2 cells for 42 of these sequences as well as 7 sequences that overlap linkage-disequilibrium blocks defined by lead SNPs from pharmacogenomic GWAS studies, revealing 15/42 and 4/7 to be functional enhancers, respectively. A common African haplotype in one of these enhancers in the GSTA locus was found to exhibit potential rifampin hypersensitivity. Combined, our results further suggest that enhancers are the predominant targets of rifampin-induced PXR activation, provide a genome-wide catalog of PXR targets and serve as a model for the identification of drug-responsive regulatory elements.


Current Opinion in Genetics & Development | 2013

A large-scale view of the evolution of amniote development: insights from somitogenesis in reptiles

Kenro Kusumi; Catherine M. May; Walter L. Eckalbar

Uncovering the genetic and developmental changes that generate morphological diversity is one of the longstanding challenges in biology. The developmental process generating the spine, one of the defining features of vertebrates, constitutes one of these core questions. The vertebral column is patterned in early development through the formation of segments, called somites, regulated by gene networks collectively called the segmentation clock. While previous studies of somite development in amniotes have focused almost exclusively on the mouse and chick model systems, the growing availability of genomic sequences in other taxa has opened up the possibility of comparative developmental studies in nontraditional reptilian models, such as the anole lizard, alligator, and snake. These studies have identified conserved features of the segmentation clock, but they have also challenged previous assumptions and identified divergence in the genetic networks. Ongoing work will help to elucidate which of these morphological changes may be explained by divergences in development in amniote evolution.


PLOS Genetics | 2016

Bat Accelerated Regions Identify a Bat Forelimb Specific Enhancer in the HoxD Locus.

Betty M. Booker; Tara Friedrich; Mandy K. Mason; Julia E. VanderMeer; Jingjing Zhao; Walter L. Eckalbar; Malcolm Logan; Nicola Illing; Katherine S. Pollard; Nadav Ahituv

The molecular events leading to the development of the bat wing remain largely unknown, and are thought to be caused, in part, by changes in gene expression during limb development. These expression changes could be instigated by variations in gene regulatory enhancers. Here, we used a comparative genomics approach to identify regions that evolved rapidly in the bat ancestor, but are highly conserved in other vertebrates. We discovered 166 bat accelerated regions (BARs) that overlap H3K27ac and p300 ChIP-seq peaks in developing mouse limbs. Using a mouse enhancer assay, we show that five Myotis lucifugus BARs drive gene expression in the developing mouse limb, with the majority showing differential enhancer activity compared to the mouse orthologous BAR sequences. These include BAR116, which is located telomeric to the HoxD cluster and had robust forelimb expression for the M. lucifugus sequence and no activity for the mouse sequence at embryonic day 12.5. Developing limb expression analysis of Hoxd10-Hoxd13 in Miniopterus natalensis bats showed a high-forelimb weak-hindlimb expression for Hoxd10-Hoxd11, similar to the expression trend observed for M. lucifugus BAR116 in mice, suggesting that it could be involved in the regulation of the bat HoxD complex. Combined, our results highlight novel regulatory regions that could be instrumental for the morphological differences leading to the development of the bat wing.


PLOS Genetics | 2016

Genomic Characterization of Metformin Hepatic Response

Marcelo R. Luizon; Walter L. Eckalbar; Yao Wang; Stacy L. Jones; Robin P. Smith; Megan Laurance; Lawrence Lin; Paul J. Gallins; Amy S. Etheridge; Fred A. Wright; Yi Hui Zhou; Cliona Molony; Federico Innocenti; Sook Wah Yee; Kathleen M. Giacomini; Nadav Ahituv

Metformin is used as a first-line therapy for type 2 diabetes (T2D) and prescribed for numerous other diseases. However, its mechanism of action in the liver has yet to be characterized in a systematic manner. To comprehensively identify genes and regulatory elements associated with metformin treatment, we carried out RNA-seq and ChIP-seq (H3K27ac, H3K27me3) on primary human hepatocytes from the same donor treated with vehicle control, metformin or metformin and compound C, an AMP-activated protein kinase (AMPK) inhibitor (allowing to identify AMPK-independent pathways). We identified thousands of metformin responsive AMPK-dependent and AMPK-independent differentially expressed genes and regulatory elements. We functionally validated several elements for metformin-induced promoter and enhancer activity. These include an enhancer in an ataxia telangiectasia mutated (ATM) intron that has SNPs in linkage disequilibrium with a metformin treatment response GWAS lead SNP (rs11212617) that showed increased enhancer activity for the associated haplotype. Expression quantitative trait locus (eQTL) liver analysis and CRISPR activation suggest that this enhancer could be regulating ATM, which has a known role in AMPK activation, and potentially also EXPH5 and DDX10, its neighboring genes. Using ChIP-seq and siRNA knockdown, we further show that activating transcription factor 3 (ATF3), our top metformin upregulated AMPK-dependent gene, could have an important role in gluconeogenesis repression. Our findings provide a genome-wide representation of metformin hepatic response, highlight important sequences that could be associated with interindividual variability in glycemic response to metformin and identify novel T2D treatment candidates.

Collaboration


Dive into the Walter L. Eckalbar's collaboration.

Top Co-Authors

Avatar

Kenro Kusumi

Arizona State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nadav Ahituv

University of California

View shared research outputs
Top Co-Authors

Avatar

Alan Rawls

Arizona State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John C. Eckalbar

California State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge