Try a new search

Format these results:

Searched for:

person:cuddas01

in-biosketch:yes

Total Results:

44


Nickel exposure induces persistent mesenchymal phenotype in human lung epithelial cells through epigenetic activation of ZEB1

Jose, Cynthia C; Jagannathan, Lakshmanan; Tanwar, Vinay Singh; Zhang, Xiaoru; Zang, Chongzhi; Cuddapah, Suresh
Nickel (Ni) is an environmental and occupational carcinogen, and exposure to Ni is associated with lung and nasal cancers in humans. Furthermore, Ni exposure is implicated in several lung diseases including chronic inflammatory airway diseases, asthma and fibrosis. However, the mutagenic potential of Ni is low and does not correlate with its potent toxicity and carcinogenicity. Therefore, mechanisms underlying Ni exposure-associated diseases remain poorly understood. Since the health risks of environmental exposures often continue post exposure, understanding the exposure effects that persist after the termination of exposure could provide mechanistic insights into diseases. By examining the persistent effects of Ni exposure, we report that Ni induces epithelial-mesenchymal transition (EMT) and that the mesenchymal phenotype remains irreversible even after the termination of exposure. Ni-induced EMT was dependent on the irreversible upregulation of ZEB1, an EMT master regulator, via resolution of its promoter bivalency. ZEB1, upon activation, downregulated its repressors as well as the cell-cell adhesion molecule, E-cadherin, resulting in the cells undergoing EMT and switching to persistent mesenchymal status. ZEB1 depletion in cells exposed to Ni attenuated Ni-induced EMT. Moreover, Ni exposure did not induce EMT in ZEB1-depleted cells. Activation of EMT, during which the epithelial cells lose cell-cell adhesion and become migratory and invasive, plays a major role in asthma, fibrosis, and cancer and metastasis, lung diseases associated with Ni exposure. Therefore, our finding of irreversible epigenetic activation of ZEB1 by Ni exposure and the acquisition of persistent mesenchymal phenotype would have important implications in understanding Ni-induced diseases.
PMCID:5930076
PMID: 29528143
ISSN: 1098-2744
CID: 2994122

Identification of a unique gene expression signature in mercury and 2,3,7,8-tetrachlorodibenzo-p-dioxin co-exposed cells

Jagannathan, Lakshmanan; Jose, Cynthia C; Tanwar, Vinay Singh; Bhattacharya, Sudin; Cuddapah, Suresh
Mercury (Hg) and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) are major environmental contaminants that commonly co-occur in the environment. Both Hg and TCDD are associated with a number of human diseases including cancers. While the individual toxicological effects of Hg and TCDD have been extensively investigated, studies on co-exposure are limited to a few genes and pathways. Therefore, a significant knowledge gap exists in the understanding of the deleterious effects of co-exposure to Hg and TCDD. Due to the prevalence of Hg and TCDD co-contamination in the environment and the major human health hazards they pose, it is important to obtain a fuller understanding of genome-wide effects of Hg and TCDD co-exposure. In this study, by performing a comprehensive transcriptomic analysis of human bronchial epithelial cells (BEAS-2B) exposed to Hg and TCDD individually and in combination, we have uncovered a subset of genes with altered expression only in the co-exposed cells. We also identified the additive as well as antagonistic effects of Hg and TCDD on gene expression. Moreover, we found that co-exposure impacted several biological and disease processes not affected by Hg or TCDD individually. Our studies show that the consequences of Hg and TCDD co-exposure on the transcriptional program and biological processes could be substantially different from single exposures, thus providing new insights into the co-exposure-specific pathogenic processes.
PMCID:5648359
PMID: 29057067
ISSN: 2045-452x
CID: 2756762

Rapid Recall Ability of Memory T cells is Encoded in their Epigenome

Barski, Artem; Cuddapah, Suresh; Kartashov, Andrey V; Liu, Chong; Imamichi, Hiromi; Yang, Wenjing; Peng, Weiqun; Lane, H Clifford; Zhao, Keji
Even though T-cell receptor (TCR) stimulation together with co-stimulation is sufficient for the activation of both naive and memory T cells, the memory cells are capable of producing lineage specific cytokines much more rapidly than the naive cells. The mechanisms behind this rapid recall response of the memory cells are still not completely understood. Here, we performed epigenetic profiling of human resting naive, central and effector memory T cells using ChIP-Seq and found that unlike the naive cells, the regulatory elements of the cytokine genes in the memory T cells are marked by activating histone modifications even in the resting state. Therefore, the ability to induce expression of rapid recall genes upon activation is associated with the deposition of positive histone modifications during memory T cell differentiation. We propose a model of T cell memory, in which immunological memory state is encoded epigenetically, through poising and transcriptional memory.
PMCID:5215294
PMID: 28054639
ISSN: 2045-2322
CID: 2386792

Lineage relationship of CD8 T cell subsets is revealed by progressive changes in the epigenetic landscape

Crompton, Joseph G; Narayanan, Manikandan; Cuddapah, Suresh; Roychoudhuri, Rahul; Ji, Yun; Yang, Wenjing; Patel, Shashank J; Sukumar, Madhusudhanan; Palmer, Douglas C; Peng, Weiqun; Wang, Ena; Marincola, Francesco M; Klebanoff, Christopher A; Zhao, Keji; Tsang, John S; Gattinoni, Luca; Restifo, Nicholas P
To better elucidate epigenetic mechanisms that correlate with the dynamic gene expression program observed upon T-cell differentiation, we investigated the genomic landscape of histone modifications in naive and memory CD8+ T cells. Using a ChIP-Seq approach coupled with global gene expression profiling, we generated genome-wide histone H3 lysine 4 (H3K4me3) and H3 lysine 27 (H3K27me3) trimethylation maps in naive, T memory stem cells, central memory cells, and effector memory cells in order to gain insight into how histone architecture is remodeled during T cell differentiation. We show that H3K4me3 histone modifications are associated with activation of genes, while H3K27me3 is negatively correlated with gene expression at canonical loci and enhancers associated with T-cell metabolism, effector function, and memory. Our results also reveal histone modifications and gene expression signatures that distinguish the recently identified T memory stem cells from other CD8+ T-cell subsets. Taken together, our results suggest that CD8+ lymphocytes undergo chromatin remodeling in a progressive fashion. These findings have major implications for our understanding of peripheral T-cell ontogeny and the formation of immunological memory.Cellular & Molecular Immunology advance online publication, 27 April 2015; doi:10.1038/cmi.2015.032.
PMCID:4947817
PMID: 25914936
ISSN: 2042-0226
CID: 1556902

Nuclear Factor kappaB1/RelA Mediates Inflammation in Human Lung Epithelial Cells at Atmospheric Oxygen Levels

Jagannathan, Lakshmanan; Jose, Cynthia C; Arita, Adriana; Kluz, Thomas; Sun, Hong; Zhang, Xiaoru; Yao, Yixin; Kartashov, Andrey V; Barski, Artem; Costa, Max; Cuddapah, Suresh
Oxygen levels range from 2-9% in vivo. Atmospheric O2 levels (21%) are known to induce cell proliferation defects and cellular senescence in primary cell cultures. However, the mechanistic basis of the deleterious effects of higher O2 levels is not fully understood. On the other hand, immortalized cells including cancer cell lines, which evade cellular senescence are normally cultured at 21% O2 and the effects of higher O2 on these cells are understudied. Here we addressed this problem by culturing immortalized human bronchial epithelial (BEAS-2B) cells at ambient atmospheric, 21% O2 and lower, 10% O2 . Our results show increased inflammatory response at 21% O2 but not at 10% O2 . We found higher RelA binding at the NF-kappaB1/RelA target gene promoters as well as upregulation of several pro-inflammatory cytokines in cells cultured at 21% O2 . RelA knockdown prevented the upregulation of the pro-inflammatory cytokines at 21% O2 , suggesting NF-kappaB1/RelA as a major mediator of inflammatory response in cells cultured at 21% O2 . Interestingly, unlike the 21% O2 cultured cells, exposure of 10% O2 cultured cells to H2 O2 did not elicit inflammatory response, suggesting increased ability to tolerate oxidative stress in cells cultured at lower O2 levels
PMCID:4845657
PMID: 26588041
ISSN: 1097-4652
CID: 1848872

Oxidative stress under ambient and physiological oxygen tension in tissue culture

Jagannathan, Lakshmanan; Cuddapah, Suresh; Costa, Max
Oxygen (O2) levels range from 2-9% in vivo. However, cell culture experiments are performed at atmospheric O2 levels (21%). Oxidative stress due to generation of reactive oxygen species (ROS) in cells cultured at higher than physiological levels is implicated in multitude of deleterious effects including DNA damage, genomic instability and senescence. In addition, oxidative stress activates redox sensitive transcription factors related to inflammatory signaling and apoptotic signaling. Furthermore, several chromatin-modifying enzymes are affected by ROS, potentially impacting epigenetic regulation of gene expression. While primary cells are cultured at lower O2 levels due to their inability to grow at higher O2, the immortalized cells, which display no such apparent growth difficulties, are typically cultured at 21% O2. This review will provide an overview of issues associated with increased oxygen levels in in vitro cell culture and point out the benefits of using lower levels of oxygen tension even for immortalized cells.
PMCID:4809260
PMID: 27034917
ISSN: 2198-641x
CID: 2059342

Epigenetic dysregulation by nickel through repressive chromatin domain disruption

Jose, Cynthia C; Xu, Beisi; Jagannathan, Lakshmanan; Trac, Candi; Mallela, Ramya K; Hattori, Takamitsu; Lai, Darson; Koide, Shohei; Schones, Dustin E; Cuddapah, Suresh
Investigations into the genomic landscape of histone modifications in heterochromatic regions have revealed histone H3 lysine 9 dimethylation (H3K9me2) to be important for differentiation and maintaining cell identity. H3K9me2 is associated with gene silencing and is organized into large repressive domains that exist in close proximity to active genes, indicating the importance of maintenance of proper domain structure. Here we show that nickel, a nonmutagenic environmental carcinogen, disrupted H3K9me2 domains, resulting in the spreading of H3K9me2 into active regions, which was associated with gene silencing. We found weak CCCTC-binding factor (CTCF)-binding sites and reduced CTCF binding at the Ni-disrupted H3K9me2 domain boundaries, suggesting a loss of CTCF-mediated insulation function as a potential reason for domain disruption and spreading. We furthermore show that euchromatin islands, local regions of active chromatin within large H3K9me2 domains, can protect genes from H3K9me2-spreading-associated gene silencing. These results have major implications in understanding H3K9me2 dynamics and the consequences of chromatin domain disruption during pathogenesis.
PMCID:4210008
PMID: 25246589
ISSN: 0027-8424
CID: 1259292

Insulators recruit histone methyltransferase dMes4 to regulate chromatin of flanking genes

Lhoumaud, Priscillia; Hennion, Magali; Gamot, Adrien; Cuddapah, Suresh; Queille, Sophie; Liang, Jun; Micas, Gael; Morillon, Pauline; Urbach, Serge; Bouchez, Olivier; Severac, Dany; Emberly, Eldon; Zhao, Keji; Cuvier, Olivier
Chromosomal domains in Drosophila are marked by the insulator-binding proteins (IBPs) dCTCF/Beaf32 and cofactors that participate in regulating long-range interactions. Chromosomal borders are further enriched in specific histone modifications, yet the role of histone modifiers and nucleosome dynamics in this context remains largely unknown. Here, we show that IBP depletion impairs nucleosome dynamics specifically at the promoters and coding sequence of genes flanked by IBP binding sites. Biochemical purification identifies the H3K36 histone methyltransferase NSD/dMes-4 as a novel IBP cofactor, which specifically co-regulates the chromatin accessibility of hundreds of genes flanked by dCTCF/Beaf32. NSD/dMes-4 presets chromatin before the recruitment of transcriptional activators including DREF that triggers Set2/Hypb-dependent H3K36 trimethylation, nucleosome positioning, and RNA splicing. Our results unveil a model for how IBPs regulate nucleosome dynamics and gene expression through NSD/dMes-4, which may regulate H3K27me3 spreading. Our data uncover how IBPs dynamically regulate chromatin organization depending on distinct cofactors.
PMCID:4198054
PMID: 24916307
ISSN: 0261-4189
CID: 1033612

Chromatin immunoprecipitation indirect peaks highlight long-range interactions of insulator proteins and Pol II pausing

Liang, Jun; Lacroix, Laurent; Gamot, Adrien; Cuddapah, Suresh; Queille, Sophie; Lhoumaud, Priscillia; Lepetit, Pierre; Martin, Pascal G P; Vogelmann, Jutta; Court, Franck; Hennion, Magali; Micas, Gael; Urbach, Serge; Bouchez, Olivier; Nollmann, Marcelo; Zhao, Keji; Emberly, Eldon; Cuvier, Olivier
Eukaryotic chromosomes are partitioned into topologically associating domains (TADs) that are demarcated by distinct insulator-binding proteins (IBPs) in Drosophila. Whether IBPs regulate specific long-range contacts and how this may impact gene expression remains unclear. Here we identify "indirect peaks" of multiple IBPs that represent their distant sites of interactions through long-range contacts. Indirect peaks depend on protein-protein interactions among multiple IBPs and their common cofactors, including CP190, as confirmed by high-resolution analyses of long-range contacts. Mutant IBPs unable to interact with CP190 impair long-range contacts as well as the expression of hundreds of distant genes that are specifically flanked by indirect peaks. Regulation of distant genes strongly correlates with RNAPII pausing, highlighting how this key transcriptional stage may trap insulator-based long-range interactions. Our data illustrate how indirect peaks may decipher gene regulatory networks through specific long-range interactions.
PMCID:4198380
PMID: 24486021
ISSN: 1097-2765
CID: 978582

Acute depletion of Tet1-dependent 5-hydroxymethylcytosine levels impairs LIF/Stat3 signaling and results in loss of embryonic stem cell identity

Freudenberg, Johannes M; Ghosh, Swati; Lackford, Brad L; Yellaboina, Sailu; Zheng, Xiaofeng; Li, Ruifang; Cuddapah, Suresh; Wade, Paul A; Hu, Guang; Jothi, Raja
The TET family of FE(II) and 2-oxoglutarate-dependent enzymes (Tet1/2/3) promote DNA demethylation by converting 5-methylcytosine to 5-hydroxymethylcytosine (5hmC), which they further oxidize into 5-formylcytosine and 5-carboxylcytosine. Tet1 is robustly expressed in mouse embryonic stem cells (mESCs) and has been implicated in mESC maintenance. Here we demonstrate that, unlike genetic deletion, RNAi-mediated depletion of Tet1 in mESCs led to a significant reduction in 5hmC and loss of mESC identity. The differentiation phenotype due to Tet1 depletion positively correlated with the extent of 5hmC loss. Meta-analyses of genomic data sets suggested interaction between Tet1 and leukemia inhibitory factor (LIF) signaling. LIF signaling is known to promote self-renewal and pluripotency in mESCs partly by opposing MAPK/ERK-mediated differentiation. Withdrawal of LIF leads to differentiation of mESCs. We discovered that Tet1 depletion impaired LIF-dependent Stat3-mediated gene activation by affecting Stat3's ability to bind to its target sites on chromatin. Nanog overexpression or inhibition of MAPK/ERK signaling, both known to maintain mESCs in the absence of LIF, rescued Tet1 depletion, further supporting the dependence of LIF/Stat3 signaling on Tet1. These data support the conclusion that analysis of mESCs in the hours/days immediately following efficient Tet1 depletion reveals Tet1's normal physiological role in maintaining the pluripotent state that may be subject to homeostatic compensation in genetic models.
PMCID:3333871
PMID: 22210859
ISSN: 0305-1048
CID: 165603