Network


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

Hotspot


Dive into the research topics where Qingdong Ke is active.

Publication


Featured researches published by Qingdong Ke.


Molecular Pharmacology | 2006

Hypoxia-Inducible Factor-1 (HIF-1)

Qingdong Ke; Max Costa

Adaptation to low oxygen tension (hypoxia) in cells and tissues leads to the transcriptional induction of a series of genes that participate in angiogenesis, iron metabolism, glucose metabolism, and cell proliferation/survival. The primary factor mediating this response is the hypoxia-inducible factor-1 (HIF-1), an oxygen-sensitive transcriptional activator. HIF-1 consists of a constitutively expressed subunit HIF-1β and an oxygen-regulated subunit HIF-1α (or its paralogs HIF-2α and HIF-3α). The stability and activity of the α subunit of HIF are regulated by its post-translational modifications such as hydroxylation, ubiquitination, acetylation, and phosphorylation. In normoxia, hydroxylation of two proline residues and acetylation of a lysine residue at the oxygen-dependent degradation domain (ODDD) of HIF-1α trigger its association with pVHL E3 ligase complex, leading to HIF-1α degradation via ubiquitin-proteasome pathway. In hypoxia, the HIF-1α subunit becomes stable and interacts with coactivators such as cAMP response element-binding protein binding protein/p300 and regulates the expression of target genes. Overexpression of HIF-1 has been found in various cancers, and targeting HIF-1 could represent a novel approach to cancer therapy.


Molecular and Cellular Biochemistry | 2004

Molecular mechanisms of arsenic carcinogenesis.

Chuanshu Huang; Qingdong Ke; Max Costa; Xianglin Shi

Arsenic is a metalloid compound that is widely distributed in the environment. Human exposure of this compound has been associated with increased cancer incidence. Although the exact mechanisms remain to be investigated, numerous carcinogenic pathways have been proposed. Potential carcinogenic actions for arsenic include oxidative stress, genotoxic damage, DNA repair inhibition, epigenetic events, and activation of certain signal transduction pathways leading to abberrant gene expression. In this article, we summarize current knowledge on the molecular mechanisms of arsenic carcinogenesis with an emphasis on ROS and signal transduction pathways.


Molecular and Cellular Biology | 2006

Nickel Ions Increase Histone H3 Lysine 9 Dimethylation and Induce Transgene Silencing

Haobin Chen; Qingdong Ke; Thomas Kluz; Yan Yan; Max Costa

ABSTRACT We have previously reported that carcinogenic nickel compounds decreased global histone H4 acetylation and silenced the gpt transgene in G12 Chinese hamster cells. However, the nature of this silencing is still not clear. Here, we report that nickel ion exposure increases global H3K9 mono- and dimethylation, both of which are critical marks for DNA methylation and long-term gene silencing. In contrast to the up-regulation of global H3K9 dimethylation, nickel ions decreased the expression and activity of histone H3K9 specific methyltransferase G9a. Further investigation demonstrated that nickel ions interfered with the removal of histone methylation in vivo and directly decreased the activity of a Fe(II)-2-oxoglutarate-dependent histone H3K9 demethylase in nuclear extract in vitro. These results are the first to show a histone H3K9 demethylase activity dependent on both iron and 2-oxoglutarate. Exposure to nickel ions also increased H3K9 dimethylation at the gpt locus in G12 cells and repressed the expression of the gpt transgene. An extended nickel ion exposure led to increased frequency of the gpt transgene silencing, which was readily reversed by treatment with DNA-demethylating agent 5-aza-2′-deoxycytidine. Collectively, our data strongly indicate that nickel ions induce transgene silencing by increasing histone H3K9 dimethylation, and this effect is mediated by the inhibition of H3K9 demethylation.


Carcinogenesis | 2008

Nickel compounds induce phosphorylation of histone H3 at serine 10 by activating JNK-MAPK pathway.

Qingdong Ke; Qin Li; Thomas P. Ellen; Hong Sun; Max Costa

Nickel (Ni) is a known carcinogen, although the mechanism of its carcinogenicity is not clear. Here, we provide evidence that Ni can induce phosphorylation of histone H3 at its serine 10 residue in a c-jun N-terminal kinase (JNK)/stress-activated protein kinase (SAPK)-dependent manner. Ni induces the phosphorylation of JNK, with no effect on the phosphorylation states of the extracellular signal-regulated kinase (ERK) or p38 mitogen-activated protein kinases. An inhibitor of JNK eliminated the Ni-initiated JNK-mediated induction of histone H3 phosphorylation at serine 10, whereas inhibitors specific for ERK or p38 kinases had no effect on the phosphorylation levels of histone H3 at serine 10 (P-H3S10) in Ni-treated cells. A complete loss of Ni ion-induced phosphorylation of H3S10 was observed when JNK was specifically knocked down with RNAi. These results are the first to show the specific JNK-mediated phosphorylation of histone H3 at its serine 10 residue. We show that addition of Ni to an in vitro P-H3S10 dephosphorylation reaction does not change the loss of phosphorylation in the reaction, supporting the notion that Ni causes H3S10 phosphorylation via the JNK/SAPK pathway. It is likely that modification of H3S10 is one of a growing number of epigenetic changes believed to be involved in the carcinogenesis caused by Ni.


Toxicology and Applied Pharmacology | 2008

NICKEL COMPOUNDS INDUCE HISTONE UBIQUITINATION BY INHIBITING HISTONE DEUBIQUITINATING ENZYME ACTIVITY

Qingdong Ke; Thomas P. Ellen; Max Costa

Nickel (Ni) compounds are known carcinogens but underlying mechanisms are not clear. Epigenetic changes are likely to play an important role in nickel ion carcinogenesis. Previous studies have shown epigenetic effects of nickel ions, including the loss of histone acetylation and a pronounced increase in dimethylated H3K9 in nickel-exposed cells. In this study, we demonstrated that both water-soluble and insoluble nickel compounds induce histone ubiquitination (uH2A and uH2B) in a variety of cell lines. Investigations of the mechanism by which nickel increases histone ubiquitination in cells reveal that nickel does not affect cellular levels of the substrates of this modification, i.e., ubiquitin, histones, and other non-histone ubiquitinated proteins. In vitro ubiquitination and deubiquitination assays have been developed to further investigate possible effects of nickel on enzymes responsible for histone ubiquitination. Results from the in vitro assays demonstrate that the presence of nickel did not affect the levels of ubiquitinated histones in the ubiquitinating assay. Instead, the addition of nickel significantly prevents loss of uH2A and uH2B in the deubiquitinating assay, suggesting that nickel-induced histone ubiquitination is the result of inhibition of (a) putative deubiquitinating enzyme(s). Additional supporting evidence comes from the comparison of the response to nickel ions with a known deubiquitinating enzyme inhibitor, iodoacetamide (IAA). This study is the first to demonstrate such effects of nickel ions on histone ubiquitination. It also sheds light on the possible mechanisms involved in altering the steady state of this modification. The study provides further evidence that supports the notion that nickel ions alter epigenetic homeostasis in cells, which may lead to altered programs of gene expression and carcinogenesis.


Carcinogenesis | 2009

Alterations of histone modifications by cobalt compounds

Qin Li; Qingdong Ke; Max Costa

In the present study, we examined the effects of CoCl(2) on multiple histone modifications at the global level. We found that in both human lung carcinoma A549 cells and human bronchial epithelial Beas-2B cells, exposure to CoCl(2) (>/=200 muM) for 24 h increased H3K4me3, H3K9me2, H3K9me3, H3K27me3, H3K36me3, uH2A and uH2B but decreased acetylation at histone H4 (AcH4). Further investigation demonstrated that in A549 cells, the increase in H3K4me3 and H3K27me3 by cobalt ions exposure was probably through enhancing histone methylation processes, as methionine-deficient medium blocked the induction of H3K4me3 and H3K27me3 by cobalt ions, whereas cobalt ions increased H3K9me3 and H3K36me3 by directly inhibiting JMJD2A demethylase activity in vitro, which was probably due to the competition of cobalt ions with iron for binding to the active site of JMJD2A. Furthermore, in vitro ubiquitination and deubiquitination assays revealed that the cobalt-induced histone H2A and H2B ubiquitination is the result of inhibition of deubiquitinating enzyme activity. Microarray data showed that exposed to 200 microM of CoCl(2) for 24 h, A549 cells not only increased but also decreased expression of hundreds of genes involved in different cellular functions, including tumorigenesis. This study is the first to demonstrate that cobalt ions altered epigenetic homeostasis in cells. It also sheds light on the possible mechanisms involved in cobalt-induced alteration of histone modifications, which may lead to altered programs of gene expression and carcinogenesis since cobalt at higher concentrations is a known carcinogen.


The Nutritional Biochemistry of Chromium (III) | 2007

Overview of chromium(III) toxicology

Qingdong Ke; Max Costa

Environmental exposure to chromium (Cr) is becoming an increasing concern, and certain Cr compounds are recognized as human carcinogens. Chronic use of Cr picolinate has been reported to cause nephrotoxicity in humans if ingested in excess. In addition, it has been reported that Cr picolinate consumption is associated with some acute physiological and behavioral impairment. Chromium supplements have been shown to generate reactive oxygen species (ROS) in cells. Reactive oxygen species are oxygen-containing and highly reactive molecules with unpaired electrons formed during oxidative metabolism. Considerable evidence has shown that ROS play an important role in the initiation of cellular injury, which can lead to the development of cancer. Humans who are exposed to Cr(III) picolinate or other environmentally relevant organic Cr(III) complexes humic acid complexes), are potentially accumulating high levels of Cr(III) in cells. This intracellular accumulation of Cr(III) can result in the formation of covalent bonds between Cr(III) and DNA and/or other macromolecules, causing genotoxic effects.


Carcinogenesis | 2007

NDRG1, a growth and cancer related gene : regulation of gene expression and function in normal and disease states

Thomas P. Ellen; Qingdong Ke; Ping Zhang; Max Costa


Mutation Research | 2005

Nickel carcinogenesis: Epigenetics and hypoxia signaling

Max Costa; Todd Davidson; Haobin Chen; Qingdong Ke; Ping Zhang; Yan Yan; Chuanshu Huang; Thomas Kluz


Carcinogenesis | 2006

Alterations of histone modifications and transgene silencing by nickel chloride

Qingdong Ke; Todd Davidson; Haobin Chen; Thomas Kluz; Max Costa

Collaboration


Dive into the Qingdong Ke's collaboration.

Top Co-Authors

Avatar

Max Costa

National Institute for Occupational Safety and Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bing-Hua Jiang

National Institute for Occupational Safety and Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qin Li

New York University

View shared research outputs
Researchain Logo
Decentralizing Knowledge