Network


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

Hotspot


Dive into the research topics where Jeffrey D. Steimle is active.

Publication


Featured researches published by Jeffrey D. Steimle.


PLOS Genetics | 2014

Foxf genes integrate tbx5 and hedgehog pathways in the second heart field for cardiac septation.

Andrew D. Hoffmann; Xinan Holly Yang; Ozanna Burnicka-Turek; Joshua D. Bosman; Xiaomeng Ren; Jeffrey D. Steimle; Steven A. Vokes; Andrew P. McMahon; Vladimir V. Kalinichenko; Ivan P. Moskowitz

The Second Heart Field (SHF) has been implicated in several forms of congenital heart disease (CHD), including atrioventricular septal defects (AVSDs). Identifying the SHF gene regulatory networks required for atrioventricular septation is therefore an essential goal for understanding the molecular basis of AVSDs. We defined a SHF Hedgehog-dependent gene regulatory network using whole genome transcriptional profiling and GLI-chromatin interaction studies. The Forkhead box transcription factors Foxf1a and Foxf2 were identified as SHF Hedgehog targets. Compound haploinsufficiency for Foxf1a and Foxf2 caused atrioventricular septal defects, demonstrating the biological relevance of this regulatory network. We identified a Foxf1a cis-regulatory element that bound the Hedgehog transcriptional regulators GLI1 and GLI3 and the T-box transcription factor TBX5 in vivo. GLI1 and TBX5 synergistically activated transcription from this cis-regulatory element in vitro. This enhancer drove reproducible expression in vivo in the posterior SHF, the only region where Gli1 and Tbx5 expression overlaps. Our findings implicate Foxf genes in atrioventricular septation, describe the molecular underpinnings of the genetic interaction between Hedgehog signaling and Tbx5, and establish a molecular model for the selection of the SHF gene regulatory network for cardiac septation.


PLOS Genetics | 2016

De Novo and Rare Variants at Multiple Loci Support the Oligogenic Origins of Atrioventricular Septal Heart Defects.

James R. Priest; Kazutoyo Osoegawa; Nebil Mohammed; Vivek Nanda; Ramendra K. Kundu; Kathleen Schultz; Edward J. Lammer; Santhosh Girirajan; Todd E. Scheetz; Daryl Waggott; Francois Haddad; Sushma Reddy; Daniel Bernstein; Trudy L. Burns; Jeffrey D. Steimle; Xinan Yang; Ivan P. Moskowitz; Richard P. Lifton; Debbie A. Nickerson; Michael J. Bamshad; Evan E. Eichler; Seema Mital; Val C. Sheffield; Thomas Quertermous; Bruce D. Gelb; Michael A. Portman; Euan A. Ashley

Congenital heart disease (CHD) has a complex genetic etiology, and recent studies suggest that high penetrance de novo mutations may account for only a small fraction of disease. In a multi-institutional cohort surveyed by exome sequencing, combining analysis of 987 individuals (discovery cohort of 59 affected trios and 59 control trios, and a replication cohort of 100 affected singletons and 533 unaffected singletons) we observe variation at novel and known loci related to a specific cardiac malformation the atrioventricular septal defect (AVSD). In a primary analysis, by combining developmental coexpression networks with inheritance modeling, we identify a de novo mutation in the DNA binding domain of NR1D2 (p.R175W). We show that p.R175W changes the transcriptional activity of Nr1d2 using an in vitro transactivation model in HUVEC cells. Finally, we demonstrate previously unrecognized cardiovascular malformations in the Nr1d2tm1-Dgen knockout mouse. In secondary analyses we map genetic variation to protein-interaction networks suggesting a role for two collagen genes in AVSD, which we corroborate by burden testing in a second replication cohort of 100 AVSDs and 533 controls (p = 8.37e-08). Finally, we apply a rare-disease inheritance model to identify variation in genes previously associated with CHD (ZFPM2, NSD1, NOTCH1, VCAN, and MYH6), cardiac malformations in mouse models (ADAM17, CHRD, IFT140, PTPRJ, RYR1 and ATE1), and hypomorphic alleles of genes causing syndromic CHD (EHMT1, SRCAP, BBS2, NOTCH2, and KMT2D) in 14 of 59 trios, greatly exceeding variation in control trios without CHD (p = 9.60e-06). In total, 32% of trios carried at least one putatively disease-associated variant across 19 loci,suggesting that inherited and de novo variation across a heterogeneous group of loci may contribute to disease risk.


Developmental Cell | 2016

The Cardiac TBX5 Interactome Reveals a Chromatin Remodeling Network Essential for Cardiac Septation

Lauren Waldron; Jeffrey D. Steimle; Todd M. Greco; Nicholas C. Gomez; Kerry M. Dorr; Junghun Kweon; Brenda Temple; Xinan Holly Yang; Caralynn M. Wilczewski; Ian J. Davis; Ileana M. Cristea; Ivan P. Moskowitz; Frank L. Conlon

Human mutations in the cardiac transcription factor gene TBX5 cause congenital heart disease (CHD), although the underlying mechanism is unknown. We report characterization of the endogenous TBX5 cardiac interactome and demonstrate that TBX5, long considered a transcriptional activator, interacts biochemically and genetically with the nucleosome remodeling and deacetylase (NuRD) repressor complex. Incompatible gene programs are repressed by TBX5 in the developing heart. CHD mis-sense mutations that disrupt the TBX5-NuRD interaction cause depression of a subset of repressed genes. Furthermore, the TBX5-NuRD interaction is required for heart development. Phylogenetic analysis showed that the TBX5-NuRD interaction domain evolved during early diversification of vertebrates, simultaneous with the evolution of cardiac septation. Collectively, this work defines a TBX5-NuRD interaction essential to cardiac development and the evolution of the mammalian heart, and when altered may contribute to human CHD.


Current Topics in Developmental Biology | 2017

TBX5: A Key Regulator of Heart Development

Jeffrey D. Steimle; Ivan P. Moskowitz

TBX5 is a member of the T-box transcription factor family and is primarily known for its role in cardiac and forelimb development. Human patients with dominant mutations in TBX5 are characterized by Holt-Oram syndrome, and show defects of the cardiac septa, cardiac conduction system, and the anterior forelimb. The range of cardiac defects associated with TBX5 mutations in humans suggests multiple roles for the transcription factor in cardiac development and function. Animal models demonstrate similar defects and have provided a useful platform for investigating the roles of TBX5 during embryonic development. During early cardiac development, TBX5 appears to act primarily as a transcriptional activator of genes associated with cardiomyocyte maturation and upstream of morphological signals for septation. During later cardiac development, TBX5 is required for patterning of the cardiac conduction system and maintenance of mature cardiomyocyte function. A comprehensive understanding of the integral roles of TBX5 throughout cardiac development and adult life will be critical for understanding human cardiac morphology and function.


BMC Developmental Biology | 2015

The pattern of congenital heart defects arising from reduced Tbx5 expression is altered in a Down syndrome mouse model

Renita C. Polk; Peter Gergics; Jeffrey D. Steimle; Huiqing Li; Ivan P. Moskowitz; Sally A. Camper; Roger H. Reeves

BackgroundNearly half of all individuals with Down Syndrome (DS) have some type of congenital heart defect (CHD), suggesting that DS sensitizes to CHD but does not cause it. We used a common mouse model of DS, the Ts65Dn mouse, to study the contribution of Tbx5, a known modifier of CHD, to heart defects on a trisomic backgroun. Mice that were heterozygous for a Tbx5 null allele were crossed with Ts65Dn mice. Thoraxes of progeny were fixed in 10% formalin, embedded in paraffin, and sectioned for analysis of CHD. Gene expression in embryonic hearts was examined by quantitative PCR and in situ hybridization. A TBX5 DNA binding site was verified by luciferase assays.MethodsMice that were heterozygous for a Tbx5 null allele were crossed with Ts65Dn mice. Thoraxes of progeny were fixed in 10 % formalin, embedded in paraffin, and sectioned for analysis of CHD. Gene expression in embryonic hearts was examined by quantitative PCR and in situ hybridization. A TBX5 DNA binding site was verified by luciferase assays.ResultsWe crossed mice that were heterozygous for a Tbx5 null allele with Ts65Dn mice. Mice that were trisomic and carried the Tbx5 mutation (Ts65Dn;Tbx5+/−) had a significantly increased incidence of overriding aorta compared to their euploid littermates. Ts65Dn;Tbx5+/− mice also showed reduced expression of Pitx2, a molecular marker for the left atrium. Transcript levels of the trisomic Adamts1 gene were decreased in Tbx5+/− mice compared to their euploid littermates. Evidence of a valid binding site for TBX5 upstream of the trisomic Adamts1 locus was also shown.ConclusionHaploinsufficiency of Tbx5 and trisomy affects alignment of the aorta and this effect may stem from deviations from normal left-right patterning in the heart. We have unveiled a previously unknown interaction between the Tbx5 gene and trisomy, suggesting a connection between Tbx5 and trisomic genes important during heart development.


bioRxiv | 2018

Hedgehog signaling controls progenitor differentiation timing during heart development

Megan Rowton; Andrew D. Hoffmann; Jeffrey D. Steimle; Xinan Holly Yang; Alexander Guzzetta; Sonja Lazarevic; Chul Kim; Nikita Deng; Emery Lu; Jessica Jacobs-Li; Shuhan Yu; Erika Hanson; Carlos Perez-Cervantes; Sunny Sun Kin Chan; Kohta Ikegami; Daniel J. Garry; Michael Kyba; Ivan P. Moskowitz

Heterochrony, defined as differences in the timing of developmental processes, impacts organ development, homeostasis, and regeneration. The molecular basis of heterochrony in mammalian tissues is poorly understood. We report that Hedgehog signaling activates a heterochronic pathway that controls differentiation timing in multiple lineages. A differentiation trajectory from second heart field cardiac progenitors to first heart field cardiomyocytes was identified by single-cell transcriptional profiling in mouse embryos. A survey of developmental signaling pathways revealed specific enrichment for Hedgehog signaling targets in cardiac progenitors. Removal of Hh signaling caused loss of progenitor and precocious cardiomyocyte differentiation gene expression in the second heart field in vivo. Introduction of active Hh signaling to mESC-derived progenitors, modelled by transient expression of the Hh-dependent transcription factor GLI1, delayed differentiation in cardiac and neural lineages in vitro. A shared GLI1-dependent network in both cardiac and neural progenitors was enriched with FOX family transcription factors. FOXF1, a GLI1 target, was sufficient to delay onset of the cardiomyocyte differentiation program in progenitors, by epigenetic repression of cardiomyocyte-specific enhancers. Removal of active Hh signaling or Foxf1 expression from second heart field progenitors caused precocious cardiac differentiation in vivo, establishing a mechanism for resultant Congenital Heart Disease. Together, these studies suggest that Hedgehog signaling directly activates a gene regulatory network that functions as a heterochronic switch to control differentiation timing across developmental lineages.A balance between progenitor cell maintenance and differentiation is tightly regulated during embryonic development and adult tissue homeostasis. Hedgehog (Hh) signaling is known to function within the progenitor cell populations of several developing and regenerative organs, and in cancers with resident progenitor subpopulations. Here we provide evidence that Hh signaling dictates differentiation timing by promoting progenitor status and inhibiting differentiation during mammalian heart morphogenesis. Removal of active Hh signaling from cardiac progenitors in vivo caused increased cardiomyocyte differentiation gene expression, precocious differentiation, and Congenital Heart Disease (CHD). Modeling active Hh signaling through expression of the activating Hh transcription factor (TF), GLI1, in cardiac progenitors enabled the maintenance of a progenitor-specific regulatory network and delayed onset of the cardiomyocyte differentiation program. Progenitor-specific, but not differentiated cardiomyocyte-specific, accessible chromatin regions were enriched for GLI binding motifs. Furthermore, GLI1 expression promoted a global shift of chromatin accessibility towards progenitor-like, and away from cardiomyocyte-like, profiles at distal regulatory elements. The shift from active to repressive GLI TF abundance comprised a molecular switch that determined the activity patterns of progenitor-specific regulatory elements in vitro and in vivo. Thus, Hh signaling modulates a GLI switch at progenitor enhancers to activate progenitor gene expression and inhibit premature differentiation, thereby determining cardiac progenitor differentiation timing. This provides a novel molecular paradigm for progenitor maintenance by signal-dependent TFs with implications for organ development, regenerative potential, and cancer.


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

Evolutionarily conserved Tbx5–Wnt2/2b pathway orchestrates cardiopulmonary development

Jeffrey D. Steimle; Scott A. Rankin; Christopher E. Slagle; Jenna Bekeny; Ariel B. Rydeen; Sunny Sun Kin Chan; Junghun Kweon; Xinan Yang; Kohta Ikegami; Rangarajan D. Nadadur; Megan Rowton; Andrew D. Hoffmann; Sonja Lazarevic; William Thomas; Erin A. T. Boyle Anderson; Marko E. Horb; Luis Luna-Zurita; Robert K. Ho; Michael Kyba; Bjarke Jensen; Aaron M. Zorn; Frank L. Conlon; Ivan P. Moskowitz

Significance In the 20 years since the discovery of the genetic link between the transcription factor TBX5 and congenital heart defects, few direct targets of TBX5 in cardiac morphogenesis have been identified. In this work, we demonstrate that TBX5 directly regulates canonical Wnt ligands required for initiation of lung development. Lung endoderm forms a Hedgehog signaling source required for morphogenesis of both the lungs and the cardiac inflow septum. Our work expands the role of TBX5 to include a non–cell-autonomous component for atrial septation. We find the mesoderm–endoderm–mesoderm signaling loop initiated by TBX5 is evolutionarily conserved from amphibians to mammals. This work suggests that the evolutionary origin of lungs may have involved the recruitment of cardiac TBX5. Codevelopment of the lungs and heart underlies key evolutionary innovations in the transition to terrestrial life. Cardiac specializations that support pulmonary circulation, including the atrial septum, are generated by second heart field (SHF) cardiopulmonary progenitors (CPPs). It has been presumed that transcription factors required in the SHF for cardiac septation, e.g., Tbx5, directly drive a cardiac morphogenesis gene-regulatory network. Here, we report instead that TBX5 directly drives Wnt ligands to initiate a bidirectional signaling loop between cardiopulmonary mesoderm and the foregut endoderm for endodermal pulmonary specification and, subsequently, atrial septation. We show that Tbx5 is required for pulmonary specification in mice and amphibians but not for swim bladder development in zebrafish. TBX5 is non–cell-autonomously required for pulmonary endoderm specification by directly driving Wnt2 and Wnt2b expression in cardiopulmonary mesoderm. TBX5 ChIP-sequencing identified cis-regulatory elements at Wnt2 sufficient for endogenous Wnt2 expression domains in vivo and required for Wnt2 expression in precardiac mesoderm in vitro. Tbx5 cooperated with Shh signaling to drive Wnt2b expression for lung morphogenesis. Tbx5 haploinsufficiency in mice, a model of Holt–Oram syndrome, caused a quantitative decrement of mesodermal-to-endodermal Wnt signaling and subsequent endodermal-to-mesodermal Shh signaling required for cardiac morphogenesis. Thus, Tbx5 initiates a mesoderm–endoderm–mesoderm signaling loop in lunged vertebrates that provides a molecular basis for the coevolution of pulmonary and cardiac structures required for terrestrial life.


Human Molecular Genetics | 2016

Cilia gene mutations cause atrioventricular septal defects by multiple mechanisms

Ozanna Burnicka-Turek; Jeffrey D. Steimle; Wenhui Huang; Lindsay Felker; Anna Kamp; Junghun Kweon; Michael A. Peterson; Roger H. Reeves; Cheryl L. Maslen; Peter J. Gruber; Xinan Yang; Jay Shendure; Ivan P. Moskowitz


Circulation | 2018

Abstract 10344: Transcriptional Patterning of the Cardiac Conduction System Prevents Arrhythmias

Ozanna Burnicka-Turek; Michael Broman; Jeffrey D. Steimle; Bastiaan J. Boukens; Kohta Ikegami; Yun Qiao; David E. Arnolds; Rangarajan D Nadadaru; Xinan Yang; Vickas V Patel; Marcelo A. Nobrega; Michael J. Rust; Igor R. Efimov; Ivan P. Moskowitz


PLOS Genetics | 2016

Correction: Foxf Genes Integrate Tbx5 and Hedgehog Pathways in the Second Heart Field for Cardiac Septation.

Andrew D. Hoffmann; Xinan Holly Yang; Ozanna Burnicka-Turek; Joshua D. Bosman; Xiaomeng Ren; Linglin Xie; Jeffrey D. Steimle; Steven A. Vokes; Andrew P. McMahon; Vladimir V. Kalinichenko; Ivan P. Moskowitz

Collaboration


Dive into the Jeffrey D. Steimle's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew P. McMahon

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Frank L. Conlon

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Igor R. Efimov

George Washington University

View shared research outputs
Researchain Logo
Decentralizing Knowledge