Network


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

Hotspot


Dive into the research topics where Isabelle N. King is active.

Publication


Featured researches published by Isabelle N. King.


Nature | 2005

Mutations in NOTCH1 cause aortic valve disease

Vidu Garg; Alecia N. Muth; Joshua F. Ransom; Marie K. Schluterman; Robert L. Barnes; Isabelle N. King; Paul Grossfeld; Deepak Srivastava

Calcification of the aortic valve is the third leading cause of heart disease in adults. The incidence increases with age, and it is often associated with a bicuspid aortic valve present in 1–2% of the population. Despite the frequency, neither the mechanisms of valve calcification nor the developmental origin of a two, rather than three, leaflet aortic valve is known. Here, we show that mutations in the signalling and transcriptional regulator NOTCH1 cause a spectrum of developmental aortic valve anomalies and severe valve calcification in non-syndromic autosomal-dominant human pedigrees. Consistent with the valve calcification phenotype, Notch1 transcripts were most abundant in the developing aortic valve of mice, and Notch1 repressed the activity of Runx2, a central transcriptional regulator of osteoblast cell fate. The hairy-related family of transcriptional repressors (Hrt), which are activated by Notch1 signalling, physically interacted with Runx2 and repressed Runx2 transcriptional activity independent of histone deacetylase activity. These results suggest that NOTCH1 mutations cause an early developmental defect in the aortic valve and a later de-repression of calcium deposition that causes progressive aortic valve disease.


Nature | 2003

GATA4 mutations cause human congenital heart defects and reveal an interaction with TBX5

Vidu Garg; Irfan S. Kathiriya; Robert Barnes; Marie K. Schluterman; Isabelle N. King; Cheryl A. Butler; Caryn Rothrock; Reenu S. Eapen; Kayoko Hirayama-Yamada; Kunitaka Joo; Rumiko Matsuoka; Jonathan C. Cohen; Deepak Srivastava

Congenital heart defects (CHDs) are the most common developmental anomaly and are the leading non-infectious cause of mortality in newborns. Only one causative gene, NKX2-5, has been identified through genetic linkage analysis of pedigrees with non-syndromic CHDs. Here, we show that isolated cardiac septal defects in a large pedigree were linked to chromosome 8p22-23. A heterozygous G296S missense mutation of GATA4, a transcription factor essential for heart formation, was found in all available affected family members but not in any control individuals. This mutation resulted in diminished DNA-binding affinity and transcriptional activity of Gata4. Furthermore, the Gata4 mutation abrogated a physical interaction between Gata4 and TBX5, a T-box protein responsible for a subset of syndromic cardiac septal defects. Conversely, interaction of Gata4 and TBX5 was disrupted by specific human TBX5 missense mutations that cause similar cardiac septal defects. In a second family, we identified a frame-shift mutation of GATA4 (E359del) that was transcriptionally inactive and segregated with cardiac septal defects. These results implicate GATA4 as a genetic cause of human cardiac septal defects, perhaps through its interaction with TBX5.


Nature Cell Biology | 2011

Notch post-translationally regulates β-catenin protein in stem and progenitor cells

Chulan Kwon; Paul Cheng; Isabelle N. King; Peter Andersen; Lincoln T. Shenje; Vishal Nigam; Deepak Srivastava

Cellular decisions of self-renewal or differentiation arise from integration and reciprocal titration of numerous regulatory networks. Notch and Wnt/β-catenin signalling often intersect in stem and progenitor cells and regulate each other transcriptionally. The biological outcome of signalling through each pathway often depends on the context and timing as cells progress through stages of differentiation. Here, we show that membrane-bound Notch physically associates with unphosphorylated (active) β-catenin in stem and colon cancer cells and negatively regulates post-translational accumulation of active β-catenin protein. Notch-dependent regulation of β-catenin protein did not require ligand-dependent membrane cleavage of Notch or the glycogen synthase kinase- 3β-dependent activity of the β-catenin destruction complex. It did, however, require the endocytic adaptor protein Numb and lysosomal activity. This study reveals a previously unrecognized function of Notch in negatively titrating active β-catenin protein levels in stem and progenitor cells.


Journal of Cell Biology | 2011

Tinman/Nkx2-5 acts via miR-1 and upstream of Cdc42 to regulate heart function across species

Li Qian; Joshua D. Wythe; Jiandong Liu; Jerome Cartry; Georg Vogler; Bhagyalaxmi Mohapatra; Robyn Otway; Yu Huang; Isabelle N. King; Marjorie Maillet; Yi Zheng; Timothy Crawley; Ouarda Taghli-Lamallem; Christopher Semsarian; Sally L. Dunwoodie; David S. Winlaw; Richard P. Harvey; Diane Fatkin; Jeffrey A. Towbin; Jeffery D. Molkentin; Deepak Srivastava; Karen Ocorr; Benoit G. Bruneau; Rolf Bodmer

Cdc42 regulates cardiac function in mice and flies downstream of a conserved Tinman/Nkx2-5–miR-1 signaling network.


Journal of Biological Chemistry | 2014

The RNA Binding Protein TDP-43 Selectively Disrupts MicroRNA-1/206 Incorporation into the RNA-Induced Silencing Complex

Isabelle N. King; Valeria Yartseva; Donaldo Salas; Abhishek Kumar; Amy Heidersbach; D. Michael Ando; Nancy R. Stallings; Jeffrey L. Elliott; Deepak Srivastava; Kathryn N. Ivey

Background: Regulation of microRNA activity independent of processing and biogenesis has not been demonstrated. Results: The RNA-binding protein, TDP-43, interacts with mature miR-1/miR-206, limiting their RNA-induced silencing complex (RISC) association and activity. Conclusion: RNA-binding proteins can selectively control microRNA activity by disrupting RISC incorporation. Significance: This is the first known microRNA-protein interaction that controls microRNA activity independent of processing. MicroRNA (miRNA) maturation is regulated by interaction of particular miRNA precursors with specific RNA-binding proteins. Following their biogenesis, mature miRNAs are incorporated into the RNA-induced silencing complex (RISC) where they interact with mRNAs to negatively regulate protein production. However, little is known about how mature miRNAs are regulated at the level of their activity. To address this, we screened for proteins differentially bound to the mature form of the miR-1 or miR-133 miRNA families. These muscle-enriched, co-transcribed miRNA pairs cooperate to suppress smooth muscle gene expression in the heart. However, they also have opposing roles, with the miR-1 family, composed of miR-1 and miR-206, promoting myogenic differentiation, whereas miR-133 maintains the progenitor state. Here, we describe a physical interaction between TDP-43, an RNA-binding protein that forms aggregates in the neuromuscular disease, amyotrophic lateral sclerosis, and the miR-1, but not miR-133, family. Deficiency of the TDP-43 Drosophila ortholog enhanced dmiR-1 activity in vivo. In mammalian cells, TDP-43 limited the activity of both miR-1 and miR-206, but not the miR-133 family, by disrupting their RISC association. Consistent with TDP-43 dampening miR-1/206 activity, protein levels of the miR-1/206 targets, IGF-1 and HDAC4, were elevated in TDP-43 transgenic mouse muscle. This occurred without corresponding Igf-1 or Hdac4 mRNA increases and despite higher miR-1 and miR-206 expression. Our findings reveal that TDP-43 negatively regulates the activity of the miR-1 family of miRNAs by limiting their bioavailability for RISC loading and suggest a processing-independent mechanism for differential regulation of miRNA activity.


Developmental Cell | 2011

A genome-wide screen reveals a role for microRNA-1 in modulating cardiac cell polarity.

Isabelle N. King; Li Qian; Jianping Liang; Yu Huang; Joseph T.C. Shieh; Chulan Kwon; Deepak Srivastava

Many molecular pathways involved in heart disease have their roots in evolutionarily ancient developmental programs that depend critically on gene dosage and timing. MicroRNAs (miRNAs) modulate gene dosage posttranscriptionally, and among these, the muscle-specific miR-1 is particularly important for developing and maintaining somatic/skeletal and cardiac muscle. To identify pathways regulated by miR-1, we performed a forward genetic screen in Drosophila using wing-vein patterning as a biological assay. We identified several unexpected genes that genetically interacted with dmiR-1, one of which was kayak, encodes a developmentally regulated transcription factor. Additional studies directed at this genetic relationship revealed a previously unappreciated function of dmiR-1 in regulating the polarity of cardiac progenitor cells. The mammalian ortholog of kayak, c-Fos, was dysregulated in hearts of gain- or loss-of-function miR-1 mutant mice in a stress-dependent manner. These findings illustrate the power of Drosophila-based screens to find points of intersection between miRNAs and conserved pathways in mammals.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Vertebrate heart growth is regulated by functional antagonism between Gridlock and Gata5

Haibo Jia; Isabelle N. King; Sameer S. Chopra; Haiyan Wan; Terri T. Ni; Charlie Jiang; Xiaoqun Guan; Samuel A. Wells; Deepak Srivastava; Tao P. Zhong

Embryonic organs attain their final dimensions through the generation of proper cell number and size, but the control mechanisms remain obscure. Here, we establish Gridlock (Grl), a Hairy-related basic helix–loop–helix (bHLH) transcription factor, as a negative regulator of cardiomyocyte proliferative growth in zebrafish embryos. Mutations in grl cause an increase in expression of a group of immediate-early growth genes, myocardial genes, and development of hyperplastic hearts. Conversely, cardiomyocytes with augmented Grl activity have diminished cell volume and fail to divide, resulting in a marked reduction in heart size. Both bHLH domain and carboxyl region are required for Grl negative control of myocardial proliferative growth. These Grl-induced cardiac effects are counterbalanced by the transcriptional activator Gata5 but not Gata4, which promotes cardiomyocyte expansion in the embryo. Biochemical analyses show that Grl forms a complex with Gata5 through the carboxyl region and can repress Gata5-mediated transcription via the bHLH domain. Hence, our studies suggest that Grl regulates embryonic heart growth via opposing Gata5, at least in part through their protein interactions in modulating gene expression.


Journal of Biological Chemistry | 2008

A rare human sequence variant reveals myocardin autoinhibition.

Joshua F. Ransom; Isabelle N. King; Vidu Garg; Deepak Srivastava

Myocardin (MYOCD) is a transcriptional co-activator that promotes cardiac or smooth muscle gene programs through its interaction with myocyte-enhancing factor (MEF2) or serum-response factor (SRF). Isoforms of MYOCD with a truncated amino terminus show increased activity when compared with those with the full-length amino terminus, but how this is achieved remains unknown. We identified a rare human sequence variation in MYOCD in a patient with congenital heart disease that resulted in a missense mutation at codon 259 (K259R). This variation created a hypomorphic cardiac isoform with impaired SRF binding and transactivation capacity but did not impair the smooth muscle isoform of MYOCD, which lacks the amino terminus. Consistent with differential effects of the amino terminus on the K259R mutation, we found that the cardiac-specific amino terminus acted in an autoinhibitory fashion to bind MYOCD via specific negatively charged residues and thereby repressed SRF-dependent MYOCD activity. This effect was exaggerated in the MYOCD-K259R mutant. The amino terminus was sufficient to impair MYOCD-dependent fibroblast conversion into smooth muscle cells as well as cardiomyocyte hypertrophy. These findings identify a novel mechanism that regulates levels of MYOCD-dependent activation of the SRF genetic program differentially in cardiac and smooth muscle.


Development | 2017

The E3 ubiquitin ligase Nedd4/Nedd4L is directly regulated by microRNA 1.

Jun-yi Zhu; Amy Heidersbach; Irfan S. Kathiriya; Bayardo I. Garay; Kathryn N. Ivey; Deepak Srivastava; Zhe Han; Isabelle N. King

miR-1 is a small noncoding RNA molecule that modulates gene expression in heart and skeletal muscle. Loss of Drosophila miR-1 produces defects in somatic muscle and embryonic heart development, which have been partly attributed to miR-1 directly targeting Delta to decrease Notch signaling. Here, we show that overexpression of miR-1 in the fly wing can paradoxically increase Notch activity independently of its effects on Delta. Analyses of potential miR-1 targets revealed that miR-1 directly regulates the 3′UTR of the E3 ubiquitin ligase Nedd4. Analysis of embryonic and adult fly heart revealed that the Nedd4 protein regulates heart development in Drosophila. Larval fly hearts overexpressing miR-1 have profound defects in actin filament organization that are partially rescued by concurrent overexpression of Nedd4. These results indicate that miR-1 and Nedd4 act together in the formation and actin-dependent patterning of the fly heart. Importantly, we have found that the biochemical and genetic relationship between miR-1 and the mammalian ortholog Nedd4-like (Nedd4l) is evolutionarily conserved in the mammalian heart, potentially indicating a role for Nedd4L in mammalian postnatal maturation. Thus, miR-1-mediated regulation of Nedd4/Nedd4L expression may serve to broadly modulate the trafficking or degradation of Nedd4/Nedd4L substrates in the heart. Summary: The cardiac-enriched microRNA miR-1 negatively regulates the E3 ubiquitin ligase Nedd4L, which ubiquitylates many proteins and controls heart development in Drosophila.


Journal of Biological Chemistry | 2004

Hairy-related Transcription Factors Inhibit GATA-dependent Cardiac Gene Expression through a Signal-responsive Mechanism

Irfan S. Kathiriya; Isabelle N. King; Masao Murakami; Masayo Nakagawa; John M. Astle; Kelly A. Gardner; Robert D. Gerard; Eric N. Olson; Deepak Srivastava; Osamu Nakagawa

Collaboration


Dive into the Isabelle N. King's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Irfan S. Kathiriya

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chulan Kwon

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Vidu Garg

Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeffrey L. Elliott

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge