Network


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

Hotspot


Dive into the research topics where Haley O. Tucker is active.

Publication


Featured researches published by Haley O. Tucker.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Dendritic cell fate is determined by BCL11A

Gregory C. Ippolito; Joseph D. Dekker; Yui Hsi Wang; Bum Kyu Lee; Arthur L. Shaffer; Jian Lin; Jason K Wall; Baeck Seung Lee; Louis M. Staudt; Yong Jun Liu; Vishwanath R. Iyer; Haley O. Tucker

Significance This work demonstrates a key role of the B lymphocyte transcription factor BCL11A in dendritic cell (DC) development. Two major DC subsets—the plasmacytoid DC (pDC) and the conventional DC (cDC)—are believed to arise from a shared precursor called the common DC progenitor (CDP). Potential precursor differences between cDC and pDC generation might nevertheless remain to be elucidated. Here, we show that mutant mice can generate CDPs and cDCs in the absence of BCL11A, whereas pDCs (and also B cells) are abolished. This study also identifies and validates BCL11A target genes using a variety of techniques, and provides a molecular model for BCL11A activity in the B lymphocyte and pDC lineages. The plasmacytoid dendritic cell (pDC) is vital to the coordinated action of innate and adaptive immunity. pDC development has not been unequivocally traced, nor has its transcriptional regulatory network been fully clarified. Here we confirm an essential requirement for the BCL11A transcription factor in fetal pDC development, and demonstrate this lineage-specific requirement in the adult organism. Furthermore, we identify BCL11A gene targets and provide a molecular mechanism for its action in pDC commitment. Embryonic germ-line deletion of Bcl11a revealed an absolute cellular, molecular, and functional absence of pDCs in fetal mice. In adults, deletion of Bcl11a in hematopoietic stem cells resulted in perturbed yet continued generation of progenitors, loss of downstream pDC and B-cell lineages, and persisting myeloid, conventional dendritic, and T-cell lineages. Challenge with virus resulted in a marked reduction of antiviral response in conditionally deleted adults. Genome-wide analyses of BCL11A DNA binding and expression revealed that BCL11A regulates transcription of E2-2 and other pDC differentiation modulators, including ID2 and MTG16. Our results identify BCL11A as an essential, lineage-specific factor that regulates pDC development, supporting a model wherein differentiation into pDCs represents a primed “default” pathway for common dendritic cell progenitors.


Neuron | 2015

Spinal Locomotor Circuits Develop Using Hierarchical Rules Based on Motorneuron Position and Identity

Christopher A. Hinckley; William A. Alaynick; Benjamin W. Gallarda; Marito Hayashi; Kathryn L. Hilde; Shawn P. Driscoll; Joseph D. Dekker; Haley O. Tucker; Tatyana O. Sharpee; Samuel L. Pfaff

The coordination of multi-muscle movements originates in the circuitry that regulates the firing patterns of spinal motorneurons. Sensory neurons rely on the musculotopic organization of motorneurons to establish orderly connections, prompting us to examine whether the intraspinal circuitry that coordinates motor activity likewise uses cell position as an internal wiring reference. We generated a motorneuron-specific GCaMP6f mouse line and employed two-photon imaging to monitor the activity of lumbar motorneurons. We show that the central pattern generator neural network coordinately drives rhythmic columnar-specific motorneuron bursts at distinct phases of the locomotor cycle. Using multiple genetic strategies to perturb the subtype identity and orderly position of motorneurons, we found that neurons retained their rhythmic activity-but cell position was decoupled from the normal phasing pattern underlying flexion and extension. These findings suggest a hierarchical basis of motor circuit formation that relies on increasingly stringent matching of neuronal identity and position.


PLOS Genetics | 2014

An integrated cell purification and genomics strategy reveals multiple regulators of pancreas development.

Cecil M. Benitez; Kun Qu; Takuya Sugiyama; Philip T. Pauerstein; Yinghua Liu; Jennifer Tsai; Xueying Gu; Amar Ghodasara; H. Efsun Arda; Jiajing Zhang; Joseph D. Dekker; Haley O. Tucker; Howard Y. Chang; Seung K. Kim

The regulatory logic underlying global transcriptional programs controlling development of visceral organs like the pancreas remains undiscovered. Here, we profiled gene expression in 12 purified populations of fetal and adult pancreatic epithelial cells representing crucial progenitor cell subsets, and their endocrine or exocrine progeny. Using probabilistic models to decode the general programs organizing gene expression, we identified co-expressed gene sets in cell subsets that revealed patterns and processes governing progenitor cell development, lineage specification, and endocrine cell maturation. Purification of Neurog3 mutant cells and module network analysis linked established regulators such as Neurog3 to unrecognized gene targets and roles in pancreas development. Iterative module network analysis nominated and prioritized transcriptional regulators, including diabetes risk genes. Functional validation of a subset of candidate regulators with corresponding mutant mice revealed that the transcription factors Etv1, Prdm16, Runx1t1 and Bcl11a are essential for pancreas development. Our integrated approach provides a unique framework for identifying regulatory genes and functional gene sets underlying pancreas development and associated diseases such as diabetes mellitus.


Genes & Development | 2015

FoxP1 orchestration of ASD-relevant signaling pathways in the striatum.

Daniel J. Araujo; Ashley Anderson; Stefano Berto; Wesley Runnels; Matthew Harper; Simon Ammanuel; Michael A. Rieger; Hung Chung Huang; Kacey Rajkovich; Kristofer W. Loerwald; Joseph D. Dekker; Haley O. Tucker; Joseph D. Dougherty; Jay R. Gibson; Genevieve Konopka

Mutations in the transcription factor Forkhead box p1 (FOXP1) are causative for neurodevelopmental disorders such as autism. However, the function of FOXP1 within the brain remains largely uncharacterized. Here, we identify the gene expression program regulated by FoxP1 in both human neural cells and patient-relevant heterozygous Foxp1 mouse brains. We demonstrate a role for FoxP1 in the transcriptional regulation of autism-related pathways as well as genes involved in neuronal activity. We show that Foxp1 regulates the excitability of striatal medium spiny neurons and that reduction of Foxp1 correlates with defects in ultrasonic vocalizations. Finally, we demonstrate that FoxP1 has an evolutionarily conserved role in regulating pathways involved in striatal neuron identity through gene expression studies in human neural progenitors with altered FOXP1 levels. These data support an integral role for FoxP1 in regulating signaling pathways vulnerable in autism and the specific regulation of striatal pathways important for vocal communication.


Developmental Biology | 2015

Foxp1/2/4 regulate endochondral ossification as a suppresser complex

Haixia Zhao; Wenrong Zhou; Zhengju Yao; Yong Wan; Jingjing Cao; Lingling Zhang; Jianzhi Zhao; Hanjun Li; Rujiang Zhou; Baojie Li; Gang Wei; Zhang Z; Catherine A. French; Joseph D. Dekker; Yingzi Yang; Simon E. Fisher; Haley O. Tucker; Xizhi Guo

Osteoblast induction and differentiation in developing long bones is dynamically controlled by the opposing action of transcriptional activators and repressors. In contrast to the long list of activators that have been discovered over past decades, the network of repressors is not well-defined. Here we identify the expression of Foxp1/2/4 proteins, comprised of Forkhead-box (Fox) transcription factors of the Foxp subfamily, in both perichondrial skeletal progenitors and proliferating chondrocytes during endochondral ossification. Mice carrying loss-of-function and gain-of-function Foxp mutations had gross defects in appendicular skeleton formation. At the cellular level, over-expression of Foxp1/2/4 in chondroctyes abrogated osteoblast formation and chondrocyte hypertrophy. Conversely, single or compound deficiency of Foxp1/2/4 in skeletal progenitors or chondrocytes resulted in premature osteoblast differentiation in the perichondrium, coupled with impaired proliferation, survival, and hypertrophy of chondrocytes in the growth plate. Foxp1/2/4 and Runx2 proteins interacted in vitro and in vivo, and Foxp1/2/4 repressed Runx2 transactivation function in heterologous cells. This study establishes Foxp1/2/4 proteins as coordinators of osteogenesis and chondrocyte hypertrophy in developing long bones and suggests that a novel transcriptional repressor network involving Foxp1/2/4 may regulate Runx2 during endochondral ossification.


Genes & Development | 2014

Arid3a is essential to execution of the first cell fate decision via direct embryonic and extraembryonic transcriptional regulation

Catherine Rhee; Bum Kyu Lee; Samuel Beck; Azeen Anjum; Kendra R. Cook; Melissa Popowski; Haley O. Tucker; Jonghwan Kim

Despite their origin from the inner cell mass, embryonic stem (ES) cells undergo differentiation to the trophectoderm (TE) lineage by repression of the ES cell master regulator Oct4 or activation of the TE master regulator Caudal-type homeobox 2 (Cdx2). In contrast to the in-depth studies of ES cell self-renewal and pluripotency, few TE-specific regulators have been identified, thereby limiting our understanding of mechanisms underlying the first cell fate decision. Here we show that up-regulation and nuclear entry of AT-rich interactive domain 3a (Arid3a) drives TE-like transcriptional programs in ES cells, maintains trophoblast stem (TS) cell self-renewal, and promotes further trophoblastic differentiation both upstream and independent of Cdx2. Accordingly, Arid3a(-/-) mouse post-implantation placental development is severely impaired, resulting in early embryonic death. We provide evidence that Arid3a directly activates TE-specific and trophoblast lineage-specific genes while directly repressing pluripotency genes via differential regulation of epigenetic acetylation or deacetylation. Our results identify Arid3a as a critical regulator of TE and placental development through execution of the commitment and differentiation phases of the first cell fate decision.


Biochimica et Biophysica Acta | 2014

Lysine methyltransferase Smyd2 suppresses p53-dependent cardiomyocyte apoptosis.

Amna Sajjad; Tatyana Novoyatleva; Silvia Vergarajauregui; Christian Troidl; Ralph T. Schermuly; Haley O. Tucker; Felix B. Engel

Apoptosis, or programmed cell death, is an essential physiological process for proper embryogenesis as well as for homeostasis during aging. In addition, apoptosis is one of the major mechanisms causing cell loss in pathophysiological conditions such as heart failure. Thus, inhibition of apoptosis is an important approach for preventive and therapeutic strategies. Here we show that the histone 3 lysine 4- and lysine 36-specific methyltransferase Smyd2 acts as an endogenous antagonistic player of p53-dependent cardiomyocyte apoptosis. Smyd2 protein levels were significantly decreased in cardiomyocytes upon cobalt chloride-induced apoptosis or myocardial infarction, while p53 expression was enhanced. siRNA-mediated knockdown of Smyd2 in cultured cardiomyocytes further enhanced cobalt chloride-induced cardiomyocyte apoptosis. In contrast, Smyd2 overexpression resulted in marked methylation of p53 and prevented its accumulation as well as apoptotic cell death in an Hsp90-independent manner. Moreover, overexpression, of Smyd2, but not Smyd2Y240F lacking a methyl transferase activity, significantly rescued CoCl2-induced apoptosis in H9c2 cardioblasts. Finally, Smyd2 cardiomyocyte-specific deletion in vivo promoted apoptotic cell death upon myocardial infarction, which correlated with enhanced expression of p53 and pro-apoptotic Bax. Collectively, our data indicate Smyd2 as a cardioprotective protein by methylating p53.


Journal of Clinical Investigation | 2017

FOXP1 controls mesenchymal stem cell commitment and senescence during skeletal aging

Hanjun Li; Pei Liu; Shuqin Xu; Yinghua Li; Joseph D. Dekker; Baojie Li; Ying Fan; Zhang Z; Yang Hong; Gong Yang; Tingting Tang; Yongxin Ren; Haley O. Tucker; Zhengju Yao; Xizhi Guo

A hallmark of aged mesenchymal stem/progenitor cells (MSCs) in bone marrow is the pivot of differentiation potency from osteoblast to adipocyte coupled with a decrease in self-renewal capacity. However, how these cellular events are orchestrated in the aging progress is not fully understood. In this study, we have used molecular and genetic approaches to investigate the role of forkhead box P1 (FOXP1) in transcriptional control of MSC senescence. In bone marrow MSCs, FOXP1 expression levels declined with age in an inverse manner with those of the senescence marker p16INK4A. Conditional depletion of Foxp1 in bone marrow MSCs led to premature aging characteristics, including increased bone marrow adiposity, decreased bone mass, and impaired MSC self-renewal capacity in mice. At the molecular level, FOXP1 regulated cell-fate choice of MSCs through interactions with the CEBP&bgr;/&dgr; complex and recombination signal binding protein for immunoglobulin &kgr; J region (RBPj&kgr;), key modulators of adipogenesis and osteogenesis, respectively. Loss of p16INK4A in Foxp1-deficient MSCs partially rescued the defects in replication capacity and bone mass accrual. Promoter occupancy analyses revealed that FOXP1 directly represses transcription of p16INK4A. These results indicate that FOXP1 attenuates MSC senescence by orchestrating their cell-fate switch while maintaining their replicative capacity in a dose- and age-dependent manner.


Developmental Biology | 2016

Defective myogenesis in the absence of the muscle-specific lysine methyltransferase SMYD1.

Harika Nagandla; Suhujey Lopez; Wei Yu; Tara L. Rasmussen; Haley O. Tucker; Robert J. Schwartz; M. David Stewart

The SMYD (SET and MYND domain) family of lysine methyltransferases harbor a unique structure in which the methyltransferase (SET) domain is intervened by a zinc finger protein-protein interaction MYND domain. SMYD proteins methylate both histone and non-histone substrates and participate in diverse biological processes including transcriptional regulation, DNA repair, proliferation and apoptosis. Smyd1 is unique among the five family members in that it is specifically expressed in striated muscles. Smyd1 is critical for development of the right ventricle in mice. In zebrafish, Smyd1 is necessary for sarcomerogenesis in fast-twitch muscles. Smyd1 is expressed in the skeletal muscle lineage throughout myogenesis and in mature myofibers, shuttling from nucleus to cytosol during myoblast differentiation. Because of this expression pattern, we hypothesized that Smyd1 plays multiple roles at different stages of myogenesis. To determine the role of Smyd1 in mammalian myogenesis, we conditionally eliminated Smyd1 from the skeletal muscle lineage at the myoblast stage using Myf5(cre). Deletion of Smyd1 impaired myoblast differentiation, resulted in fewer myofibers and decreased expression of muscle-specific genes. Muscular defects were temporally restricted to the second wave of myogenesis. Thus, in addition to the previously described functions for Smyd1 in heart development and skeletal muscle sarcomerogenesis, these results point to a novel role for Smyd1 in myoblast differentiation.


Blood | 2016

Interferon-α signaling promotes embryonic HSC maturation.

Peter Geon Kim; Matthew C. Canver; Catherine Rhee; Samantha J. Ross; June V. Harriss; Ho-Chou Tu; Stuart H. Orkin; Haley O. Tucker; George Q. Daley

In the developing mouse embryo, the first hematopoietic stem cells (HSCs) arise in the aorta-gonad-mesonephros (AGM) and mature as they transit through the fetal liver (FL). Compared with FL and adult HSCs, AGM HSCs have reduced repopulation potential in irradiated adult transplant recipients but mechanisms underlying this deficiency in AGM HSCs are poorly understood. By co-expression gene network analysis, we deduced that AGM HSCs show lower levels of interferon-α (IFN-α)/Jak-Stat1-associated gene expression than FL HSCs. Treatment of AGM HSCs with IFN-α enhanced long-term hematopoietic engraftment and donor chimerism. Conversely, IFN-α receptor-deficient AGMs (Ifnαr1(-/-)), had significantly reduced donor chimerism. We identify adenine-thymine-rich interactive domain-3a (Arid3a), a factor essential for FL and B lymphopoiesis, as a key transcriptional co-regulator of IFN-α/Stat1 signaling. Arid3a occupies the genomic loci of Stat1 as well as several IFN-α effector genes, acting to regulate their expression. Accordingly, Arid3a(-/-) AGM HSCs had significantly reduced transplant potential, which was rescued by IFN-α treatment. Our results implicate the inflammatory IFN-α/Jak-Stat pathway in the developmental maturation of embryonic HSCs, whose manipulation may lead to increased potency of reprogrammed HSCs for transplantation.

Collaboration


Dive into the Haley O. Tucker's collaboration.

Top Co-Authors

Avatar

Joseph D. Dekker

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Bum Kyu Lee

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Catherine Rhee

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Gregory C. Ippolito

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Vishwanath R. Iyer

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Jonghwan Kim

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xizhi Guo

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Zhengju Yao

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Arthur L. Shaffer

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge