Network


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

Hotspot


Dive into the research topics where Taeko K. Noah is active.

Publication


Featured researches published by Taeko K. Noah.


Experimental Cell Research | 2011

Intestinal development and differentiation

Taeko K. Noah; Bridgitte Donahue; Noah F. Shroyer

In this review, we present an overview of intestinal development and cellular differentiation of the intestinal epithelium. The review is separated into two sections: Section one summarizes organogenesis of the small and large intestines, including endoderm and gut tube formation in early embryogenesis, villus morphogenesis, and crypt formation. Section two reviews cell fate specification and differentiation of each cell type within the intestinal epithelium. Growth factor and transcriptional networks that regulate these developmental processes are summarized.


Experimental Cell Research | 2010

SAM pointed domain ETS factor (SPDEF) regulates terminal differentiation and maturation of intestinal goblet cells

Taeko K. Noah; Avedis Kazanjian; Jeffrey A. Whitsett; Noah F. Shroyer

BACKGROUND AND AIMSnSPDEF (also termed PDEF or PSE) is an ETS family transcription factor that regulates gene expression in the prostate and goblet cell hyperplasia in the lung. Spdef has been reported to be expressed in the intestine. In this paper, we identify an important role for Spdef in regulating intestinal epithelial cell homeostasis and differentiation.nnnMETHODSnSPDEF expression was inhibited in colon cancer cells to determine its ability to control goblet cell gene activation. The effects of transgenic expression of Spdef on intestinal differentiation and homeostasis were determined.nnnRESULTSnIn LS174T colon cancer cells treated with Notch/gamma-secretase inhibitor to activate goblet cell gene expression, shRNAs that inhibited SPDEF also repressed expression of goblet cell genes AGR2, MUC2, RETLNB, and SPINK4. Transgenic expression of Spdef caused the expansion of intestinal goblet cells and corresponding reduction in Paneth, enteroendocrine, and absorptive enterocytes. Spdef inhibited proliferation of intestinal crypt cells without induction of apoptosis. Prolonged expression of the Spdef transgene caused a progressive reduction in the number of crypts that expressed Spdef, consistent with its inhibitory effects on cell proliferation.nnnCONCLUSIONSnSpdef was sufficient to inhibit proliferation of intestinal progenitors and induce differentiation into goblet cells; SPDEF was required for activation of goblet cell associated genes in vitro. These data support a model in which Spdef promotes terminal differentiation into goblet cells of a common goblet/Paneth progenitor.


Annual Review of Physiology | 2013

Notch in the Intestine: Regulation of Homeostasis and Pathogenesis

Taeko K. Noah; Noah F. Shroyer

The small and large intestines are tubular organs composed of several tissue types. The columnar epithelium that lines the inner surface of the intestines distinguishes the digestive physiology of each region of the intestine and consists of several distinct cell types that are rapidly and continually renewed by intestinal stem cells that reside near the base of the crypts of Lieberkühn. Notch signaling controls the fate of intestinal stem cells by regulating the expression of Hes genes and by repressing Atoh1. Alternate models of Notch pathway control of cell fate determination are presented. Roles for Notch signaling in development of the intestine, including mesenchymal and neural cells, are discussed. The oncogenic activities of Notch in colorectal cancer, as well as the tumor suppressive activities of Atoh1, are reviewed. Therapeutic targeting of the Notch pathway in colorectal cancers is discussed, along with potential caveats.


Gastroenterology | 2010

Atonal homolog 1 is required for growth and differentiation effects of Notch/γ-secretase inhibitors on normal and cancerous intestinal epithelial cells

Avedis Kazanjian; Taeko K. Noah; Douglas Brown; Jarred T. Burkart; Noah F. Shroyer

BACKGROUND & AIMSnThe atonal homolog 1 (Atoh1) transcription factor is required for intestinal secretory (goblet, Paneth, enteroendocrine) cell differentiation. Notch/gamma-secretase inhibitors (GSIs) block proliferation and induce secretory cell differentiation in the intestine. We used genetic analyses of mice to determine whether Atoh1 mediates the effects of GSIs in normal and cancerous intestinal epithelia.nnnMETHODSnWe studied mice with intestine-specific disruption of Atoh1 (Atoh1(Deltaintestine)), the adenomatosis polyposis coli (APC)(min) mutation, both mutations (Atoh1(Deltaintestine); APC(min)), or littermate controls; mice were given GSI or vehicle. Colorectal cancer (CRC) cell lines were treated with GSI or vehicle and with small hairpin RNAs to reduce ATOH1. Differentiation and homeostasis were assessed by protein, RNA, and histologic analyses.nnnRESULTSnGSIs failed to induce secretory cell differentiation or apoptosis or decrease proliferation of Atoh1-null progenitor cells, compared with wild-type cells. Exposure of APC(min) adenomas to GSIs decreased proliferation and increased secretory cell numbers in an Atoh1-dependent manner. In CRC cells treated with GSI, ATOH1 levels were correlated inversely with proliferation. ATOH1 was required for secretory cell gene expression in cell lines and in mice.nnnCONCLUSIONSnATOH1 is required for all effects of GSIs in intestinal crypts and adenomas; Notch has no unique function in intestinal progenitors and cancer cells other than to regulate ATOH1 expression. Reducing ATOH1 activity might mitigate intestinal toxicity from systemic GSI therapy for nonintestinal diseases. Among gastrointestinal malignancies, ATOH1 mediates the effects of GSIs, so ATOH1 expression levels might predict responses to these inhibitors. We propose that only the subset of CRCs that retain ATOH1 expression will respond to GSIs.


Gastroenterology | 2013

SPDEF functions as a colorectal tumor suppressor by inhibiting β-catenin activity.

Taeko K. Noah; Yuan-Hung Lo; Allison Price; Gang Chen; Eileen King; Mary Kay Washington; Bruce J. Aronow; Noah F. Shroyer

BACKGROUND & AIMSnExpression of the SAM pointed domain containing ETS transcription factor (SPDEF or prostate-derived ETS factor) is regulated by Atoh1 and is required for the differentiation of goblet and Paneth cells. SPDEF has been reported to suppress the development of breast, prostate, and colon tumors. We analyzed levels of SPDEF in colorectal tumor samples from patients and its tumor-suppressive functions in mouse models of colorectal cancer (CRC).nnnMETHODSnWe analyzed levels of SPDEF messenger RNA and protein in more than 500 human CRC samples and more than 80 nontumor controls. Spdef(-/-)and wild-type mice (controls) were either bred with Apc(Min/+) mice, or given azoxymethane (AOM) and dextran sodium sulfate (DSS), or 1,2-dimethylhydrazine and DSS, to induce colorectal tumors. Expression of Spdef also was induced transiently by administration of tetracycline to Spdef(dox-intestine) mice with established tumors, induced by the combination of AOM and DSS or by breeding with Apc(Min/+) mice. Colon tissues were collected and analyzed for tumor number, size, grade, and for cell proliferation and apoptosis. We also analyzed the effects of SPDEF expression in HCT116 and SW480 human CRC cells.nnnRESULTSnIn colorectal tumors from patients, loss of SPDEF was observed in approximately 85% of tumors and correlated with progression from normal tissue, to adenoma, to adenocarcinoma. Spdef(-/-); Apc(Min/+) mice developed approximately 3-fold more colon tumors than Spdef(+/+); Apc(Min/+) mice. Likewise, Spdef(-/-) mice developed approximately 3-fold more colon tumors than Spdef(+/+) mice after administration of AOM and DSS. After administration of 1,2-dimethylhydrazine and DSS, invasive carcinomas were observed exclusively in Spdef(-/-) mice. Conversely, expression of SPDEF was sufficient to promote cell-cycle exit in cells of established adenomas from Spdef(dox-intestine); Apc(Min/+) mice and in Spdef(dox-intestine) mice after administration of AOM + DSS. SPDEF inhibited the expression of β-catenin-target genes in mouse colon tumors, and interacted with β-catenin to block its transcriptional activity in CRC cell lines, resulting in lower levels of cyclin D1 and c-MYC.nnnCONCLUSIONSnSPDEF is a colon tumor suppressor and a candidate therapeutic target for colon adenomas and adenocarcinoma.


Stem cell reports | 2015

Activated STAT5 Confers Resistance to Intestinal Injury by Increasing Intestinal Stem Cell Proliferation and Regeneration

Shila Gilbert; Harini Nivarthi; Christopher N. Mayhew; Yuan-Hung Lo; Taeko K. Noah; Jefferson Vallance; Thomas Rülicke; Mathias Müller; Anil G. Jegga; Wenjuan Tang; Dongsheng Zhang; Michael A. Helmrath; Noah F. Shroyer; Richard Moriggl; Xiaonan Han

Summary Intestinal epithelial stem cells (IESCs) control the intestinal homeostatic response to inflammation and regeneration. The underlying mechanisms are unclear. Cytokine-STAT5 signaling regulates intestinal epithelial homeostasis and responses to injury. We link STAT5 signaling to IESC replenishment upon injury by depletion or activation of Stat5 transcription factor. We found that depletion of Stat5 led to deregulation of IESC marker expression and decreased LGR5+ IESC proliferation. STAT5-deficient mice exhibited worse intestinal histology and impaired crypt regeneration after γ-irradiation. We generated a transgenic mouse model with inducible expression of constitutively active Stat5. In contrast to Stat5 depletion, activation of STAT5 increased IESC proliferation, accelerated crypt regeneration, and conferred resistance to intestinal injury. Furthermore, ectopic activation of STAT5 in mouse or human stem cells promoted LGR5+ IESC self-renewal. Accordingly, STAT5 promotes IESC proliferation and regeneration to mitigate intestinal inflammation. STAT5 is a functional therapeutic target to improve the IESC regenerative response to gut injury.


Cellular and molecular gastroenterology and hepatology | 2017

Transcriptional Regulation by ATOH1 and its Target SPDEF in the Intestine

Yuan-Hung Lo; Eunah Chung; Zhaohui Li; Ying-Wooi Wan; Maxime M. Mahe; Min-Shan Chen; Taeko K. Noah; Kristin N. Bell; Hari Krishna Yalamanchili; Tiemo J. Klisch; Zhandong Liu; Joo-Seop Park; Noah F. Shroyer

Background & Aims The transcription factor atonal homolog 1 (ATOH1) controls the fate of intestinal progenitors downstream of the Notch signaling pathway. Intestinal progenitors that escape Notch activation express high levels of ATOH1 and commit to a secretory lineage fate, implicating ATOH1 as a gatekeeper for differentiation of intestinal epithelial cells. Although some transcription factors downstream of ATOH1, such as SPDEF, have been identified to specify differentiation and maturation of specific cell types, the bona fide transcriptional targets of ATOH1 still largely are unknown. Here, we aimed to identify ATOH1 targets and to identify transcription factors that are likely to co-regulate gene expression with ATOH1. Methods We used a combination of chromatin immunoprecipitation and messenger RNA–based high-throughput sequencing (ChIP-seq and RNA-seq), together with cell sorting and transgenic mice, to identify direct targets of ATOH1, and establish the epistatic relationship between ATOH1 and SPDEF. Results By using unbiased genome-wide approaches, we identified more than 700 genes as ATOH1 transcriptional targets in adult small intestine and colon. Ontology analysis indicated that ATOH1 directly regulates genes involved in specification and function of secretory cells. De novo motif analysis of ATOH1 targets identified SPDEF as a putative transcriptional co-regulator of ATOH1. Functional epistasis experiments in transgenic mice show that SPDEF amplifies ATOH1-dependent transcription but cannot independently initiate transcription of ATOH1 target genes. Conclusions This study unveils the direct targets of ATOH1 in the adult intestines and illuminates the transcriptional events that initiate the specification and function of intestinal secretory lineages.


Gastroenterology | 2017

SPDEF Induces Quiescence of Colorectal Cancer Cells by Changing the Transcriptional Targets of β-catenin

Yuan-Hung Lo; Taeko K. Noah; Min Shan Chen; Winnie Y. Zou; Ester Borras; Eduardo Vilar; Noah F. Shroyer

BACKGROUND & AIMSnThe canonical Wnt signaling pathway activates the transcriptional activity of β-catenin. This pathway is often activated in colorectal cancer cells, but strategies to block it in tumors have not been effective. The SAM pointed domain containing ETS transcription factor (SPDEF) suppresses formation of colon tumors by unclear mechanisms. We investigated these mechanisms and the effects of SPDEF on β-catenin activity in mouse models of colorectal cancer (CRC), CRC cellxa0lines, and mouse and human normal and cancer colonoids.nnnMETHODSnWe performed studies of Lgr5CreERT2; β-cateninexon3; Rosa26LSL-rtta-ires-EGFP; TRE-Spdef mice, which express an oncogenic form of β-catenin in Lgr5-positive ISCs upon administration of tamoxifen and SPDEF upon administration of tetracycline. CRC lines (HCT116 and SW480) were engineered to express inducible tagged SPDEF or vector (control) and subcutaneously injected into immunodeficient NSG mice. We generated SPDEF-inducible human colonoids, including a line derived from normal rectal mucosa (control) and an adenocarcinoma line derived from a patient with germline MUTYH mutation. Full-length and truncated forms of SPDEF were expressed in CRC cells; cells were assayed for β-catenin activity and studied in immunoprecipitation and chromatin immunoprecipitation assays.nnnRESULTSnExpression of SPDEF was sufficient to inhibit intestinal tumorigenesis by activated β-catenin, block tumor cell proliferation, and restrict growth of established tumors. In tumor cells with activated β -catenin, expression of SPDEF induced a quiescent state, which was reversed when SPDEF expression was stopped. In mouse and human normal and tumor-derived enteroids/colonoids, those that expressed SPDEF for 3 days were significantly smaller. SPDEF inhibited the transcriptional activity of β-catenin via a protein-protein interaction, independent of SPDEF DNA binding capacity. SPDEF disrupted β-catenin binding to TCF1 and TCF3, displacing β-catenin from enhancer regions of genes that regulate the cell cycle but not genes that regulate stem cell activities.nnnCONCLUSIONSnIn studies of mice and human CRC, we found that SPDEF induces a quiescent state in CRC cells by disrupting binding of β-catenin to TCF1 and TCF3 and regulation of genes that control the cell cycle. In this model, β-catenin activity determines the proliferation or quiescence of CRC cells based on the absence or presence of SPDEF.


Clinical & Experimental Allergy | 2017

Lipopolysaccharide suppresses IgE-mast cell-mediated reactions

Nianrong Wang; Melanie McKell; Andrew T. Dang; Amnah Yamani; Lisa Waggoner; Simone Vanoni; Taeko K. Noah; David Wu; Anna Kordowski; Jörg Köhl; Kasper Hoebe; Senad Divanovic; Simon P. Hogan

Clinical and experimental analyses have identified a central role for IgE/FcεRI/mast cells in promoting IgE‐mediated anaphylaxis. Recent data from human studies suggest that bacterial infections can alter susceptibility to anaphylaxis.


The Journal of Allergy and Clinical Immunology | 2018

Solute carrier family 9, subfamily A, member 3 (SLC9A3)/sodium-hydrogen exchanger member 3 (NHE3) dysregulation and dilated intercellular spaces in patients with eosinophilic esophagitis

Chang Zeng; Simone Vanoni; David Wu; Julie M. Caldwell; Justin C. Wheeler; Kavisha Arora; Taeko K. Noah; Lisa Waggoner; John A. Besse; Amnah Yamani; Jazib Uddin; Mark Rochman; Ting Wen; Mirna Chehade; Margaret H. Collins; Vincent A. Mukkada; Philip E. Putnam; Anjaparavanda P. Naren; Marc E. Rothenberg; Simon P. Hogan

Background: Eosinophilic esophagitis (EoE) is characterized by histopathologic modifications of esophageal tissue, including eosinophil‐rich inflammation, basal zone hyperplasia, and dilated intercellular spaces (DIS). The underlying molecular processes that drive the histopathologic features of EoE remain largely unexplored. Objective: We sought to investigate the involvement of solute carrier family 9, subfamily A, member 3 (SLC9A3) in esophageal epithelial intracellular pH (pHi) and DIS formation and the histopathologic features of EoE. Methods: We examined expression of esophageal epithelial gene networks associated with regulation of pHi in the EoE transcriptome of primary esophageal epithelial cells and an in vitro esophageal epithelial 3‐dimensional model system (EPC2‐ALI). Molecular and cellular analyses and ion transport assays were used to evaluate the expression and function of SLC9A3. Results: We identified altered expression of gene networks associated with regulation of pHi and acid‐protective mechanisms in esophageal biopsy specimens from pediatric patients with EoE (healthy subjects, n = 6; patients with EoE, n = 10). The most dysregulated gene central to regulating pHi was SLC9A3. SLC9A3 expression was increased within the basal layer of esophageal biopsy specimens from patients with EoE, and expression positively correlated with disease severity (eosinophils/high‐power field) and DIS (healthy subjects, n = 10; patients with EoE, n = 10). Analyses of esophageal epithelial cells revealed IL‐13–induced, signal transducer and activator of transcription 6–dependent SLC9A3 expression and Na+‐dependent proton secretion and that SLC9A3 activity correlated positively with DIS formation. Finally, we showed that IL‐13–mediated, Na+‐dependent proton secretion was the primary intracellular acid‐protective mechanism within the esophageal epithelium and that blockade of SLC9A3 transport abrogated IL‐13–induced DIS formation. Conclusions: SLC9A3 plays a functional role in DIS formation, and pharmacologic interventions targeting SLC9A3 function may suppress the histopathologic manifestations in patients with EoE. GRAPHICAL ABSTRACT Figure. No caption available.

Collaboration


Dive into the Taeko K. Noah's collaboration.

Top Co-Authors

Avatar

Noah F. Shroyer

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Simon P. Hogan

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

David Wu

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lisa Waggoner

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Yuan-Hung Lo

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Amnah Yamani

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Simone Vanoni

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Avedis Kazanjian

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Chang Zeng

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge