Network


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

Hotspot


Dive into the research topics where Daret K. St. Clair is active.

Publication


Featured researches published by Daret K. St. Clair.


Free Radical Biology and Medicine | 2008

ROS and p53: A versatile partnership

Bin Liu; Yumin Chen; Daret K. St. Clair

The tumor suppressor protein p53 is a redox-active transcription factor that organizes and directs cellular responses in the face of a variety of stresses that lead to genomic instability. One of the most important questions in the study of p53 is how selective transactivation of certain p53 target genes is achieved. Reactive oxygen species (ROS), generated by cells as products or by-products, can function either as signaling molecules or as cellular toxicants. Cellular generation of ROS is central to redox signaling. Recent studies have revealed that each cellular concentration and distribution of p53 has a distinct cellular function and that ROS act as both an upstream signal that triggers p53 activation and a downstream factor that mediates apoptosis. Here, we examine the newly discovered role of p53 in regulating cellular ROS generation and how ROS modulate selective transactivation of p53 target genes. The focus is on interlinks between ROS and p53.


Free Radical Biology and Medicine | 2009

Regulation of superoxide dismutase genes: Implications in disease

Lu Miao; Daret K. St. Clair

Numerous short-lived and highly reactive oxygen species (ROS) such as superoxide (O2(.-)), hydroxyl radical, and hydrogen peroxide are continuously generated in vivo. Depending upon concentration, location, and intracellular conditions, ROS can cause toxicity or act as signaling molecules. The cellular levels of ROS are controlled by antioxidant enzymes and small-molecule antioxidants. As major antioxidant enzymes, superoxide dismutases (SODs), including copper-zinc superoxide dismutase (Cu/ZnSOD), manganese superoxide dismutase, and extracellular superoxide dismutase, play a crucial role in scavenging O2(.-). This review focuses on the regulation of the sod genes coding for these enzymes, with an emphasis on the human genes. Current knowledge about sod structure and regulation is summarized and depicted as diagrams. Studies to date on genes coding for Cu/ZnSOD (sod1) are mostly focused on alterations in the coding region and their associations with amyotrophic lateral sclerosis. Evaluation of nucleotide sequences reveals that regulatory elements of the sod2 gene reside in both the noncoding and the coding region. Changes associated with sod2 lead to alterations in expression levels as well as protein function. We also discuss the structural basis for the changes in SOD expression associated with pathological conditions and where more work is needed to establish the relationship between SODs and diseases.


Neurobiology of Disease | 2006

Redox proteomics identification of oxidatively modified hippocampal proteins in mild cognitive impairment: insights into the development of Alzheimer's disease.

D. Allan Butterfield; H. Fai Poon; Daret K. St. Clair; Jeffery N. Keller; William M. Pierce; Jon B. Klein; William R. Markesbery

Mild cognitive impairment (MCI) is generally referred to the transitional zone between normal cognitive function and early dementia or clinically probable Alzheimers disease (AD). Oxidative stress plays a significant role in AD and is increased in the superior/middle temporal gyri of MCI subjects. Because AD involves hippocampal-resident memory dysfunction, we determined protein oxidation and identified the oxidized proteins in the hippocampi of MCI subjects. We found that protein oxidation is significantly increased in the hippocampi of MCI subjects when compared to age- and sex-matched controls. By using redox proteomics, we determined the oxidatively modified proteins in MCI hippocampus to be alpha-enolase (ENO1), glutamine synthetase (GLUL), pyruvate kinase M2 (PKM2) and peptidyl-prolyl cis/trans isomerase 1 (PIN1). The interacteome of these proteins revealed that these proteins functionally interact with SRC, hypoxia-inducible factor 1, plasminogen (PLG), MYC, tissue plasminogen activator (PLAT) and BCL2L1. Moreover, the interacteome indicates the functional involvement of energy metabolism, synaptic plasticity and mitogenesis/proliferation. Therefore, oxidative inactivation of ENO1, GLUL and PIN1 may alter these cellular processes and lead to the development of AD from MCI. We conclude that protein oxidation plays a significant role in the development of AD from MCI and that the oxidative inactivation of ENO1, GLUL, PKM2 and PIN1 is involved in the progression of AD from MCI. The current study provides a framework for future studies on the development of AD from MCI relevant to oxidative stress.


Cancer Cell | 2013

Loss of FBP1 by snail-mediated repression provides metabolic advantages in basal-like breast cancer

Chenfang Dong; Tingting Yuan; Yadi Wu; Yifan Wang; Teresa W.-M. Fan; Sumitra Miriyala; Yiwei Lin; Jun Yao; Jian Shi; Tiebang Kang; Pawel Lorkiewicz; Daret K. St. Clair; Mien Chie Hung; B. Mark Evers; Binhua P. Zhou

The epithelial-mesenchymal transition (EMT) enhances cancer invasiveness and confers tumor cells with cancer stem cell (CSC)-like characteristics. We show that the Snail-G9a-Dnmt1 complex, which is critical for E-cadherin promoter silencing, is also required for the promoter methylation of fructose-1,6-biphosphatase (FBP1) in basal-like breast cancer (BLBC). Loss of FBP1 induces glycolysis and results in increased glucose uptake, macromolecule biosynthesis, formation of tetrameric PKM2, and maintenance of ATP production under hypoxia. Loss of FBP1 also inhibits oxygen consumption and reactive oxygen species production by suppressing mitochondrial complex I activity; this metabolic reprogramming results in an increased CSC-like property and tumorigenicity by enhancing the interaction of β-catenin with T-cell factor. Our study indicates that the loss of FBP1 is a critical oncogenic event in EMT and BLBC.


Oncogene | 1997

Suppression of the malignant phenotype of human glioma cells by overexpression of manganese superoxide dismutase

Weixiong Zhong; Larry W. Oberley; Terry D. Oberley; Daret K. St. Clair

Manganese superoxide dismutase (MnSOD) has been previously shown to suppress the malignant phenotype of human melanoma and breast cancer cells. To test the possible role of MnSOD in glioma malignancy, MnSOD was overexpressed in wild type human glioma U118 cells and subcloned U118-9 cells by transfection of human MnSOD cDNA. The MnSOD-transfected cell lines demonstrated expression of exogenous (plasmid) MnSOD mRNA, increase in MnSOD immunoreactive protein, and a three- to eightfold increase in MnSOD enzymatic activity. The MnSOD overexpressing cell lines became less malignant as demonstrated by requiring a higher serum concentration to grow in vitro and much slower tumor growth in nude mice than the parental and neo control cell lines. These findings further support the hypothesis that MnSOD may be a tumor suppressor gene in a wide variety of human tumors.


Cancer Research | 2005

p53 Translocation to Mitochondria Precedes Its Nuclear Translocation and Targets Mitochondrial Oxidative Defense Protein-Manganese Superoxide Dismutase

Yunfeng Zhao; Luksana Chaiswing; Joyce M. Velez; Ines Batinic-Haberle; Nancy H. Colburn; Terry D. Oberley; Daret K. St. Clair

The tumor suppressor gene p53 is activated by reactive oxygen species-generating agents. After activation, p53 migrates to mitochondria and nucleus, a response that eventually leads to apoptosis, but how the two events are related is unknown. Herein, we show that p53 translocation to mitochondria precedes its translocation to nucleus in JB6 skin epidermal cells treated with the tumor promoter 12-O-tetradecanoylphorbol-13-acetate (TPA). Translocation of p53 to mitochondria occurs within 10 minutes after TPA application. In the mitochondria, p53 interacts with the primary antioxidant enzyme, manganese superoxide dismutase (MnSOD), consistent with the reduction of its superoxide scavenging activity, and a subsequent decrease of mitochondrial membrane potential. In contrast to the immediate action on mitochondria, p53 transcriptional activity in the nucleus increases at 1 hour following TPA application, accompanied by an increase in the levels of its target gene bax at 15 hours following TPA treatment. Activation of p53 transcriptional activity is preventable by application of a SOD mimetic (MnTE-2-PyP5+). Thus, p53 translocation to mitochondria and subsequent inactivation of MnSOD explains the observed mitochondrial dysfunction, which leads to transcription-dependent mechanisms of p53-induced apoptosis.


Archives of Biochemistry and Biophysics | 1987

Increase in manganese superoxide dismutase activity in the mouse heart after X-irradiation☆

Larry W. Oberley; Daret K. St. Clair; Anne P. Autor; Terry D. Oberley

Local X-irradiation of mouse heart caused a large increase in manganese superoxide dismutase activity (MnSOD) in this organ but not in copper and zinc containing superoxide dismutase (Cu-Zn SOD) activity. MnSOD induction was both dose and time dependent. Another mitochondrial enzyme, citrate synthase, was not induced by X-irradiation. The amount of immunoreactive MnSOD also increased after X-irradiation, showing that the amount of MnSOD protein increased after X-irradiation. The response to X-irradiation was found to be biphasic--with one large peak and one smaller peak of manganese superoxide dismutase activity. The effect of various inhibitors of cellular activities on these two peaks of MnSOD activity was examined. Cycloheximide, a cytosolic protein synthesis inhibitor, abolished both peaks of MnSOD activity, while chloramphenicol, a mitochondrial protein synthesis inhibitor, has no effect on either peak. Actinomycin D, a RNA-synthesis inhibitor, lowered both peaks, but had more of an effect on the second peak than on the first. In vivo protein synthesis studies using [3H]arginine showed that an increase in new protein synthesis occurred during the time period of the second peak, but did not occur during the first peak. These results are consistent with the hypothesis that MnSOD induction occurs in two peaks with the first peak due to a preformed MnSOD protein or mRNA for MnSOD and the second peak due to an increase in new protein synthesis.


Free Radical Biology and Medicine | 1994

Expression of manganese superoxide dismutase promotes cellular differentiation

Daret K. St. Clair; Terry D. Oberley; Kenneth E. Muse; William H. St. Clair

Manganese superoxide dismutase (MnSOD) is a nuclear encoded mitochondrial matrix enzyme that scavenges toxic superoxide radicals. It has been shown that increased generation of reactive oxygen species is associated with the differentiation of microorganisms. To test the hypothesis that the ability of mitochondrial superoxide dismutase to neutralize a cellular hyperoxidant state is important for differentiation of mammalian cells, we examined the effect of transfection of MnSOD into mouse embryo fibroblasts on cellular differentiation. C3H10T1/2 cells served as a model for differentiation because these cells can be triggered to differentiate into myoblasts, adipocytes, and chondrocytes by treatment with 5-azacytidine. In this report, myoblast differentiation was defined by the presence of multinucleated cells, appearance of Z-bands, and expression of actin and desmin in the differentiated cells. Transfection of MnSOD gene was found to greatly enhance differentiation of C3H10T1/2 cells into myoblasts by 5-azacytidine. This result identifies MnSOD as an important factor for cell differentiation and supports a role for reactive oxygen species in the process of cellular differentiation.


Free Radical Biology and Medicine | 2012

Manganese superoxide dismutase regulation and cancer

Sanjit K. Dhar; Daret K. St. Clair

Mitochondria are the power plants of the eukaryotic cell and the integrators of many metabolic activities and signaling pathways important for the life and death of a cell. Normal aerobic cells use oxidative phosphorylation to generate ATP, which supplies energy for metabolism. To drive ATP production, electrons are passed along the electron transport chain, with some leaking as superoxide during the process. It is estimated that, during normal respiration, intramitochondrial superoxide concentrations can reach 10⁻¹² M. This extremely high level of endogenous superoxide production dictates that mitochondria are equipped with antioxidant systems that prevent consequential oxidative injury to mitochondria and maintain normal mitochondrial functions. The major antioxidant enzyme that scavenges superoxide anion radical in mitochondria is manganese superoxide dismutase (MnSOD). Extensive studies on MnSOD have demonstrated that MnSOD plays a critical role in the development and progression of cancer. Many human cancer cells harbor low levels of MnSOD proteins and enzymatic activity, whereas some cancer cells possess high levels of MnSOD expression and activity. This apparent variation in MnSOD level among cancer cells suggests that differential regulation of MnSOD exists in cancer cells and that this regulation may be linked to the type and stage of cancer development. This review summarizes current knowledge of the relationship between MnSOD levels and cancer with a focus on the mechanisms regulating MnSOD expression.


Oncogene | 1999

Mutations in the promoter reveal a cause for the reduced expression of the human manganese superoxide dismutase gene in cancer cells

Yong Xu; Anuradha Krishnan; X. Steven Wan; Hideyuki J. Majima; Che-Chung Yeh; Gabriele Ludewig; Edward J. Kasarskis; Daret K. St. Clair

Manganese superoxide dismutase (MnSOD) has been shown to play an important role in preventing the development of cancer. MnSOD activity is reduced in many transformed cells and tumor tissues. We previously showed that the reduced level of MnSOD activity in cancer cells was not due to a defect in the primary structure of MnSOD protein, but rather was due to defects in gene expression. To elucidate the cause for the reduced expression of human MnSOD in cancer, we investigated the nucleotide sequence in the regulatory region of the MnSOD gene in a normal human cell line and various human tumor cell lines. A DNA fragment spanning 3.4 kb 5′ flanking region of the MnSOD gene isolated from a normal human genomic DNA library was used to determine the DNA sequence of MnSOD promoter. PCR primers were used for amplification of the 3.4 kb 5′ flanking region of the human MnSOD gene in cancer cells. Sequence analysis identified three heterozygous mutations in the proximal region of the promoter in five human tumor cell lines. These mutations, clustered around the GC-rich region of the human MnSOD promoter, change the binding pattern of AP-2 and lead to a reduction in transcription activity using a luciferase reporter assay system. These results suggest that the reduced level of MnSOD expression in some tumor cells is, at least in part, due to a defect in the DNA sequence of the promoter region.

Collaboration


Dive into the Daret K. St. Clair's collaboration.

Top Co-Authors

Avatar

Yong Xu

University of Kentucky

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Terry D. Oberley

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Teresa Noel

University of Kentucky

View shared research outputs
Top Co-Authors

Avatar

Mary Vore

University of Kentucky

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge