Network


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

Hotspot


Dive into the research topics where Preeti Ramadoss is active.

Publication


Featured researches published by Preeti Ramadoss.


Molecular Endocrinology | 2009

STAT3 Targets the Regulatory Regions of Gluconeogenic Genes in Vivo

Preeti Ramadoss; Nathan E. Unger-Smith; Francis S. Lam; Anthony N. Hollenberg

The regulation of expression of gluconeogenic genes including glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxykinase (PEPCK) in the liver plays an important role in glucose homeostasis, because aberrant expression of these genes contributes to the development of type 2 diabetes. Previous reports demonstrate that signal transducer and activator of transcription 3 (STAT3) plays a key role in regulating gluconeogenic gene expression, but the mechanism remains unclear. Herein we demonstrate that phosphorylated STAT3 is required for repression of G6Pase expression by IL-6 in both HepG2 cells and mouse liver. Interestingly, PEPCK expression is regulated by STAT3 independent of IL-6 activation. Using in vivo chromatin immunoprecipitation, we demonstrate that STAT3 binds to the promoters of the G6Pase, PEPCK, and suppressor of cytokine signaling (SOCS)3 genes, and its recruitment increases at the G6Pase and SOCS3 promoters with IL-6 treatment. Whereas persistent recruitment of RNA polymerase II is seen on the SOCS3 promoter, consistent with its induction by IL-6, a decrease in polymerase II recruitment and histone H4 acetylation is seen at the G6Pase promoter with IL-6 treatment. Thus STAT3 mediates negative regulation of hepatic gluconeogenic gene expression in vivo by interacting with regulatory regions of these genes.


Journal of Biological Chemistry | 2014

Novel Mechanism of Positive versus Negative Regulation by Thyroid Hormone Receptor β1 (TRβ1) Identified by Genome-wide Profiling of Binding Sites in Mouse Liver

Preeti Ramadoss; Brian J. Abraham; Linus T.-Y. Tsai; Yiming Zhou; Ricardo H. Costa-e-Sousa; Felix Ye; Martin Bilban; Keji Zhao; Anthony N. Hollenberg

Background: Examining the TRβ1 cistrome in mouse liver is critical to understanding thyroid hormone signaling. Results: Novel mechanisms of positive versus negative regulation by biotinylated TRβ1 were identified. Conclusion: TRβ1 regulates transcription by changes in relative binding and use of preferred binding motifs. Significance: This study demonstrates that a mechanism other than differential co-regulator recruitment is involved in transcriptional regulation by TRβ1 Triiodothyronine (T3) regulates key metabolic processes in the liver through the thyroid hormone receptor, TRβ1. However, the number of known target genes directly regulated by TRβ1 is limited, and the mechanisms by which positive and especially negative transcriptional regulation occur are not well understood. To characterize the TRβ1 cistrome in vivo, we expressed a biotinylated TRβ1 in hypo- and hyperthyroid mouse livers, used ChIP-seq to identify genomic TRβ1 targets, and correlated these data with gene expression changes. As with other nuclear receptors, the majority of TRβ1 binding sites were not in proximal promoters but in the gene body of known genes. Remarkably, T3 can dictate changes in TRβ1 binding, with strong correlation to T3-induced gene expression changes, suggesting that differential TRβ1 binding regulates transcriptional outcome. Additionally, DR-4 and DR-0 motifs were significantly enriched at binding sites where T3 induced an increase or decrease in TRβ1 binding, respectively, leading to either positive or negative regulation by T3. Taken together, the results of this study provide new insights into the mechanisms of transcriptional regulation by TRβ1 in vivo.


Cell Metabolism | 2011

NPY and MC4R Signaling Regulate Thyroid Hormone Levels during Fasting through Both Central and Peripheral Pathways

Kristen R. Vella; Preeti Ramadoss; Francis S. Lam; Jamie C. Harris; Felix Ye; Paul D. Same; Nicholas F. O'Neill; Eleftheria Maratos-Flier; Anthony N. Hollenberg

Fasting-induced suppression of the hypothalamic-pituitary-thyroid (HPT) axis is an adaptive response to decrease energy expenditure during food deprivation. Previous studies demonstrate that leptin communicates nutritional status to the HPT axis through thyrotropin-releasing hormone (TRH) in the paraventricular nucleus (PVN) of the hypothalamus. Leptin targets TRH neurons either directly or indirectly via the arcuate nucleus through pro-opiomelanocortin (POMC) and agouti-related peptide/neuropeptide Y (AgRP/NPY) neurons. To evaluate the role of these pathways in vivo, we developed double knockout mice that lack both the melanocortin 4 receptor (MC4R) and NPY. We show that NPY is required for fasting-induced suppression of Trh expression in the PVN. However, both MC4R and NPY are required for activation of hepatic pathways that metabolize T(4) during the fasting response. Thus, these signaling pathways play a key role in the communication of fasting signals to reduce thyroid hormone levels both centrally and through a peripheral hepatic circuit.


Molecular and Cellular Biology | 2014

Thyroid hormone signaling in vivo requires a balance between coactivators and corepressors.

Kristen R. Vella; Preeti Ramadoss; Ricardo H. Costa-e-Sousa; Inna Astapova; Felix Ye; Kaila A. Holtz; Jamie C. Harris; Anthony N. Hollenberg

ABSTRACT Resistance to thyroid hormone (RTH), a human syndrome, is characterized by high thyroid hormone (TH) and thyroid-stimulating hormone (TSH) levels. Mice with mutations in the thyroid hormone receptor beta (TRβ) gene that cannot bind steroid receptor coactivator 1 (SRC-1) and Src-1−/− mice both have phenotypes similar to that of RTH. Conversely, mice expressing a mutant nuclear corepressor 1 (Ncor1) allele that cannot interact with TRβ, termed NCoRΔID, have low TH levels and normal TSH. We hypothesized that Src-1−/− mice have RTH due to unopposed corepressor action. To test this, we crossed NCoRΔID and Src-1−/− mice to create mice deficient for coregulator action in all cell types. Remarkably, NCoRΔID/ΔID Src-1−/− mice have normal TH and TSH levels and are triiodothryonine (T3) sensitive at the level of the pituitary. Although absence of SRC-1 prevented T3 activation of key hepatic gene targets, NCoRΔID/ΔID Src-1−/− mice reacquired hepatic T3 sensitivity. Using in vivo chromatin immunoprecipitation assays (ChIP) for the related coactivator SRC-2, we found enhanced SRC-2 recruitment to TR-binding regions of genes in NCoRΔID/ΔID Src-1−/− mice, suggesting that SRC-2 is responsible for T3 sensitivity in the absence of NCoR1 and SRC-1. Thus, T3 targets require a critical balance between NCoR1 and SRC-1. Furthermore, replacement of NCoR1 with NCoRΔID corrects RTH in Src-1−/− mice through increased SRC-2 recruitment to T3 target genes.


Endocrinology | 2016

Liganded thyroid hormone receptors transactivate the DNA methyltransferase 3a gene in mouse neuronal cells

Yasuhiro Kyono; Arasakumar Subramani; Preeti Ramadoss; Anthony N. Hollenberg; Ronald M. Bonett; Robert J. Denver

Thyroid hormone (T3) is essential for proper neurological development. The hormone, bound to its receptors, regulates gene transcription in part by modulating posttranslational modifications of histones. Methylation of DNA, which is established by the de novo DNA methyltransferase (DNMT)3a and DNMT3b, and maintained by DNMT1 is another epigenetic modification influencing gene transcription. The expression of Dnmt3a, but not other Dnmt genes, increases in mouse brain in parallel with the postnatal rise in plasma [T3]. We found that treatment of the mouse neuroblastoma cell line Neuro2a[TRβ1] with T3 caused rapid induction of Dnmt3a mRNA, which was resistant to protein synthesis inhibition, supporting that it is a direct T3-response gene. Injection of T3 into postnatal day 6 mice increased Dnmt3a mRNA in the brain by 1 hour. Analysis of two chromatin immunoprecipitation-sequencing datasets, and targeted analyses using chromatin immunoprecipitation, transfection-reporter assays, and in vitro DNA binding identified 2 functional T3-response elements (TREs) at the mouse Dnmt3a locus located +30.3 and +49.3 kb from the transcription start site. Thyroid hormone receptors associated with both of these regions in mouse brain chromatin, but with only 1 (+30.3 kb) in Neuro2a[TRβ1] cells. Deletion of the +30.3-kb TRE using CRISPR/Cas9 genome editing eliminated or strongly reduced the Dnmt3a mRNA response to T3. Bioinformatics analysis showed that both TREs are highly conserved among eutherian mammals. Thyroid regulation of Dnmt3a may be an evolutionarily conserved mechanism for modulating global changes in DNA methylation during postnatal neurological development.


Cell Metabolism | 2017

Hypothalamic-Pituitary Axis Regulates Hydrogen Sulfide Production

Christopher Hine; Hyo Jeong Kim; Yan Zhu; Eylul Harputlugil; Alban Longchamp; Marina Souza Matos; Preeti Ramadoss; Kevin Bauerle; Lear E. Brace; John M. Asara; C. Keith Ozaki; Sheue Yann Cheng; Subhankar Singha; Kyo Han Ahn; Alec C. Kimmelman; ffolliott M. Fisher; Pavlos Pissios; Dominic J. Withers; Colin Selman; Rui Wang; Kelvin Yen; Valter D. Longo; Pinchas Cohen; Andrzej Bartke; John J. Kopchick; Richard A. Miller; Anthony N. Hollenberg; James R. Mitchell

Summary Decreased growth hormone (GH) and thyroid hormone (TH) signaling are associated with longevity and metabolic fitness. The mechanisms underlying these benefits are poorly understood, but may overlap with those of dietary restriction (DR), which imparts similar benefits. Recently we discovered that hydrogen sulfide (H2S) is increased upon DR and plays an essential role in mediating DR benefits across evolutionary boundaries. Here we found increased hepatic H2S production in long-lived mouse strains of reduced GH and/or TH action, and in a cell-autonomous manner upon serum withdrawal in vitro. Negative regulation of hepatic H2S production by GH and TH was additive and occurred via distinct mechanisms, namely direct transcriptional repression of the H2S-producing enzyme cystathionine γ-lyase (CGL) by TH, and substrate-level control of H2S production by GH. Mice lacking CGL failed to downregulate systemic T4 metabolism and circulating IGF-1, revealing an essential role for H2S in the regulation of key longevity-associated hormones.


Journal of Clinical Investigation | 2014

Hepatic nuclear corepressor 1 regulates cholesterol absorption through a TRβ1-governed pathway

Inna Astapova; Preeti Ramadoss; Ricardo H. Costa-e-Sousa; Felix Ye; Kaila A. Holtz; Yingxia Li; Michele W. Niepel; David E. Cohen; Anthony N. Hollenberg

Transcriptional coregulators are important components of nuclear receptor (NR) signaling machinery and provide additional mechanisms for modulation of NR activity. Expression of a mutated nuclear corepressor 1 (NCoR1) that lacks 2 NR interacting domains (NCoRΔID) in the liver leads to elevated expression of genes regulated by thyroid hormone receptor (TR) and liver X receptor (LXR), both of which control hepatic cholesterol metabolism. Here, we demonstrate that expression of NCoRΔID in mouse liver improves dietary cholesterol tolerance in an LXRα-independent manner. NCoRΔID-associated cholesterol tolerance was primarily due to diminished intestinal cholesterol absorption as the result of changes in the composition and hydrophobicity of the bile salt pool. Alterations of the bile salt pool were mediated by increased expression of genes encoding the bile acid metabolism enzymes CYP27A1 and CYP3A11 as well as canalicular bile salt pump ABCB11. We have determined that these genes are regulated by thyroid hormone and that TRβ1 is recruited to their regulatory regions. Together, these data indicate that interactions between NCoR1 and TR control a specific pathway involved in regulation of cholesterol metabolism and clearance.


Molecular and Cellular Endocrinology | 2013

Family members CREB and CREM control thyrotropin-releasing hormone (TRH) expression in the hypothalamus

Franck Chiappini; Preeti Ramadoss; Kristen R. Vella; Lucas L. Cunha; Felix Ye; Ronald C. Stuart; Eduardo A. Nillni; Anthony N. Hollenberg

Thyrotropin-releasing hormone (TRH) in the paraventricular nucleus (PVN) of the hypothalamus is regulated by thyroid hormone (TH). cAMP response element binding protein (CREB) has also been postulated to regulate TRH expression but its interaction with TH signaling in vivo is not known. To evaluate the role of CREB in TRH regulation in vivo, we deleted CREB from PVN neurons to generate the CREB1(ΔSIM1) mouse. As previously shown, loss of CREB was compensated for by an up-regulation of CREM in euthyroid CREB1(ΔSIM1) mice but TSH, T₄ and T₃ levels were normal, even though TRH mRNA levels were elevated. Interestingly, TRH mRNA expression was also increased in the PVN of CREB1(ΔSIM1) mice in the hypothyroid state but became normal when made hyperthyroid. Importantly, CREM levels were similar in CREB1(ΔSIM1) mice regardless of thyroid status, demonstrating that the regulation of TRH by T₃ in vivo likely occurs independently of the CREB/CREM family.


Diabetes | 2018

T3 Induces Both Markers of Maturation and Aging in Pancreatic β-Cells

Cristina Aguayo-Mazzucato; Terence B. Lee; Michelle Matzko; Amanda DiIenno; Habib Rezanejad; Preeti Ramadoss; Thomas S. Scanlan; Ann Marie Zavacki; P. Reed Larsen; Anthony N. Hollenberg; Clark K. Colton; Arun Sharma; Susan Bonner-Weir

Previously, we showed that thyroid hormone (TH) triiodothyronine (T3) enhanced β-cell functional maturation through induction of Mafa. High levels of T3 have been linked to decreased life span in mammals and low levels to lengthened life span, suggesting a relationship between TH and aging. Here, we show that T3 increased p16Ink4a (a β-cell senescence marker and effector) mRNA in rodent and human β-cells. The kinetics of Mafa and p16Ink4a induction suggested both genes as targets of TH via TH receptors (THRs) binding to specific response elements. Using specific agonists CO23 and GC1, we showed that p16Ink4a expression was controlled by THRA and Mafa by THRB. Using chromatin immunoprecipitation and a transient transfection yielding biotinylated THRB1 or THRA isoforms to achieve specificity, we determined that THRA isoform bound to p16Ink4a, whereas THRB1 bound to Mafa but not to p16Ink4a. On a cellular level, T3 treatment accelerated cell senescence as shown by increased number of β-cells with acidic β-galactosidase activity. Our data show that T3 can simultaneously induce both maturation (Mafa) and aging (p16Ink4a) effectors and that these dichotomous effects are mediated through different THR isoforms. These findings may be important for further improving stem cell differentiation protocols to produce functional β-cells for replacement therapies in diabetes.


Endocrine Reviews | 2011

The Nuclear Receptor Corepressor (NCoR) Controls Thyroid Hormone Sensitivity and the Set Point of the Hypothalamic-Pituitary-Thyroid Axis

Inna Astapova; Kristen R. Vella; Preeti Ramadoss; Kaila A. Holtz; Benjamin A. Rodwin; Xiao Hui Liao; Roy E. Weiss; Michael A. Rosenberg; Anthony Rosenzweig; Anthony N. Hollenberg

This article appears in Molecular Endocrinology, published January 14, 2011, 10.1210/me.2010-0462

Collaboration


Dive into the Preeti Ramadoss's collaboration.

Top Co-Authors

Avatar

Anthony N. Hollenberg

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Felix Ye

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Kristen R. Vella

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Inna Astapova

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Kaila A. Holtz

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ricardo H. Costa-e-Sousa

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Francis S. Lam

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jamie C. Harris

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Alban Longchamp

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge