Network


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

Hotspot


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

Publication


Featured researches published by D.K. St. Clair.


Neuroscience | 2008

A neuronal model of Alzheimer's disease: an insight into the mechanisms of oxidative stress-mediated mitochondrial injury.

Pradoldej Sompol; Wanida Ittarat; Jitbanjong Tangpong; Yumin Chen; I. Doubinskaia; Ines Batinic-Haberle; Hafiz Mohmmad Abdul; D.A. Butterfield; D.K. St. Clair

Alzheimers disease (AD) is associated with beta-amyloid accumulation, oxidative stress and mitochondrial dysfunction. However, the effects of genetic mutation of AD on oxidative status and mitochondrial manganese superoxide dismutase (MnSOD) production during neuronal development are unclear. To investigate the consequences of genetic mutation of AD on oxidative damages and production of MnSOD during neuronal development, we used primary neurons from new born wild-type (WT/WT) and amyloid precursor protein (APP) (NLh/NLh) and presenilin 1 (PS1) (P264L) knock-in mice (APP/PS1) which incorporated humanized mutations in the genome. Increasing levels of oxidative damages, including protein carbonyl, 4-hydroxynonenal (4-HNE) and 3-nitrotyrosine (3-NT), were accompanied by a reduction in mitochondrial membrane potential in both developing and mature APP/PS1 neurons compared with WT/WT neurons suggesting mitochondrial dysfunction under oxidative stress. Interestingly, developing APP/PS1 neurons were significantly more resistant to beta-amyloid 1-42 treatment, whereas mature APP/PS1 neurons were more vulnerable than WT/WT neurons of the same age. Consistent with the protective function of MnSOD, developing APP/PS1 neurons have increased MnSOD protein and activity, indicating an adaptive response to oxidative stress in developing neurons. In contrast, mature APP/PS1 neurons exhibited lower MnSOD levels compared with mature WT/WT neurons indicating that mature APP/PS1 neurons lost the adaptive response. Moreover, mature APP/PS1 neurons had more co-localization of MnSOD with nitrotyrosine indicating a greater inhibition of MnSOD by nitrotyrosine. Overexpression of MnSOD or addition of MnTE-2-PyP(5+) (SOD mimetic) protected against beta-amyloid-induced neuronal death and improved mitochondrial respiratory function. Together, the results demonstrate that compensatory induction of MnSOD in response to an early increase in oxidative stress protects developing neurons against beta-amyloid toxicity. However, continuing development of neurons under oxidative damage conditions may suppress the expression of MnSOD and enhance cell death in mature neurons.


The FASEB Journal | 1999

Overexpression of manganese superoxide dismutase protects against mitochondrial-initiated poly(ADP-ribose) polymerase-mediated cell death

Kelley K. Kiningham; Terry D. Oberley; Shu-Mei Lin; C. A. Mattingly; D.K. St. Clair

Mitochondria have recently been shown to serve a central role in programmed cell death. In addition, reactive oxygen species (ROS) have been implicated in cell death pathways upon treatment with a variety of agents; however, the specific cellular source of the ROS generation is unknown. We hypothesize that mitochondria‐derived free radicals play a critical role in apoptotic cell death. To directly test this hypothesis, we treated murine fibrosarcoma cell lines, which expressed a range of mitochondrial manganese superoxide dis‐mutase (MnSOD) activities, with respiratory chain inhibitors. Apoptosis was confirmed by DNA fragmentation analysis and electron microscopy. MnSOD overexpression specifically protected against cell death upon treatment with rotenone or antimycin. We examined bcl‐xL, p53 and poly(ADP‐ribose) polymerase (PARP) to identify specific cellular pathways that might contribute to the mitochondrial‐initiated ROS‐mediated cell death. Cells overexpressing Mn‐SOD contained less bcl‐xL within the mitochondria compared to control (NEO) cells, therefore excluding the role of bcl‐xL. p53 was undetectable by Western analysis and examination of the proapo‐ptotic protein bax, a p53 target gene, did not increase with treatment. Activation of caspase‐3 (CPP‐32) occurred in the NEO cells independent of cytochrome c release from the mitochondria. PARP, a target protein of CPP‐32 activity, was cleaved to a 64 kDa fragment in the NEO cells prior to generation of nucleosomal fragments. Taken together, these findings suggest that mitochondrial‐mediated ROS generation is a key event by which inhibition of respiration causes cell death, and identifies CPP‐32 and the PARP‐linked pathway as targets of mitochon‐drial‐derived ROS‐induced cell death.—Kiningham, K. K., Oberley, T. D., Lin, S.‐M., Mattingly, C. A., St. Clair, D. K. Overexpression of manganese superoxide dismutase protects against mitochondrial‐initi‐ated poly(ADP‐ribose) polymerase‐mediated cell death. FASEB J. 13, 1601–1610 (1999)


Oncogene | 2006

RelB regulates manganese superoxide dismutase gene and resistance to ionizing radiation of prostate cancer cells.

S. Josson; Yong Xu; Fang Fang; Sanjit K. Dhar; D.K. St. Clair; W. St. Clair

The relationship between NF-κB and resistance to radiation treatment in many tumor cell types has been generally well recognized. However, which members of the NF-κB family contribute to radiation resistance is unclear. In the present study, we demonstrate that RelB plays an important radioprotective role in aggressive prostate cancer cells, in part by the induction of antioxidant and antiapoptotic manganese superoxide dismutase (MnSOD) gene. RelB is both constitutively present and is inducible by radiation in aggressive prostate cancer cells. Using ectopically expressed dominant negative inhibitor, p100 mutant, and the siRNA approach, we demonstrate that selective inhibition of RelB significantly decreases the levels of MnSOD resulting in a significant increase in the sensitivity of prostate cancer cells to radiation treatment. These results demonstrate that RelB plays an important role in redox regulation of the cell and protects aggressive prostate cancer cells against radiation-induced cell death. Thus, inhibition of RelB could be a novel mechanism to radiosensitize prostate cancer.


Free Radical Biology and Medicine | 2014

Redox proteomic identification of HNE-bound mitochondrial proteins in cardiac tissues reveals a systemic effect on energy metabolism after doxorubicin treatment.

Yanming Zhao; Sumitra Miriyala; Lu Miao; Mihail I. Mitov; David M. Schnell; Sanjit K. Dhar; Jian Cai; Jon B. Klein; Rukhsana Sultana; D.A. Butterfield; Mary Vore; Ines Batinic-Haberle; Subbarao Bondada; D.K. St. Clair

Doxorubicin (DOX), one of the most effective anticancer drugs, is known to generate progressive cardiac damage, which is due, in part, to DOX-induced reactive oxygen species (ROS). The elevated ROS often induce oxidative protein modifications that result in alteration of protein functions. This study demonstrates that the level of proteins adducted by 4-hydroxy-2-nonenal (HNE), a lipid peroxidation product, is significantly increased in mouse heart mitochondria after DOX treatment. A redox proteomics method involving two-dimensional electrophoresis followed by mass spectrometry and investigation of protein databases identified several HNE-modified mitochondrial proteins, which were verified by HNE-specific immunoprecipitation in cardiac mitochondria from the DOX-treated mice. The majority of the identified proteins are related to mitochondrial energy metabolism. These include proteins in the citric acid cycle and electron transport chain. The enzymatic activities of the HNE-adducted proteins were significantly reduced in DOX-treated mice. Consistent with the decline in the function of the HNE-adducted proteins, the respiratory function of cardiac mitochondria as determined by oxygen consumption rate was also significantly reduced after DOX treatment. Treatment with Mn(III) meso-tetrakis(N-n-butoxyethylpyridinium-2-yl)porphyrin, an SOD mimic, averted the doxorubicin-induced mitochondrial dysfunctions as well as the HNE-protein adductions. Together, the results demonstrate that free radical-mediated alteration of energy metabolism is an important mechanism mediating DOX-induced cardiac injury, suggesting that metabolic intervention may represent a novel approach to preventing cardiac injury after chemotherapy.


Neuroscience | 2008

Tumor necrosis factor alpha–mediated nitric oxide production enhances manganese superoxide dismutase nitration and mitochondrial dysfunction in primary neurons: an insight into the role of glial cells

Jitbanjong Tangpong; Pradoldej Sompol; Mary Vore; W. St. Clair; D.A. Butterfield; D.K. St. Clair

Tumor necrosis factor-alpha (TNF-alpha), a ubiquitous pro-inflammatory cytokine, is an important mediator in the immune-neuroendocrine system that affects the CNS. The present study demonstrates that treatment with TNF-alpha activates microglia to increase TNF-alpha production in primary cultures of glial cells isolated from wild-type (WT) mice and mice deficient in the inducible form of nitric oxide synthase (iNOSKO). However, mitochondrial dysfunction in WT neurons occurs at lower concentrations of TNF-alpha when neurons are directly treated with TNF-alpha or co-cultured with TNF-alpha-treated microglia than iNOSKO neurons similarly treated. Immunofluorescent staining of primary neurons co-cultured with TNF-alpha-treated microglia reveals that the antioxidant enzyme in mitochondria, manganese superoxide dismutase (MnSOD), is co-localized with nitrotyrosine in WT but not in iNOSKO primary neuronal cells. Importantly, the percentage of surviving neurons is significantly reduced in WT neurons compared with iNOSKO neurons under identical treatment conditions. Together, the results suggest that TNF-alpha activates microglia to produce high levels of TNF-alpha and that production of nitric oxide (NO) in neurons is an important factor affecting MnSOD nitration and subsequent mitochondrial dysfunction.


Oncogene | 2015

Mitochondrial SOD2 regulates epithelial–mesenchymal transition and cell populations defined by differential CD44 expression

Hideaki Kinugasa; Kelly A. Whelan; Koji Tanaka; M Natsuizaka; A Long; A Guo; S Chang; Shingo Kagawa; Satish Srinivasan; Manti Guha; K Yamamoto; D.K. St. Clair; Narayan G. Avadhani; J A Diehl; Hiroshi Nakagawa

Epithelial–mesenchymal transition (EMT) promotes cancer cell invasion, metastasis and treatment failure. EMT may be activated in cancer cells by reactive oxygen species (ROS). EMT may promote conversion of a subset of cancer cells from a CD44low-CD24high (CD44L) epithelial phenotype to a CD44high-CD24−/low (CD44H) mesenchymal phenotype, the latter associated with increased malignant properties of cancer cells. ROS are required for cells undergoing EMT, although excessive ROS may induce cell death or senescence; however, little is known as to how cellular antioxidant capabilities may be regulated during EMT. Mitochondrial superoxide dismutase 2 (SOD2) is frequently overexpressed in oral and esophageal cancers. Here, we investigate mechanisms of SOD2 transcriptional regulation in EMT, as well as the functional role of this antioxidant in EMT. Using well-characterized genetically engineered oral and esophageal human epithelial cell lines coupled with RNA interference and flow cytometric approaches, we find that transforming growth factor (TGF)-β stimulates EMT, resulting in conversion of CD44L to CD44H cells, the latter of which display SOD2 upregulation. SOD2 induction in transformed keratinocytes was concurrent with suppression of TGF-β-mediated induction of both ROS and senescence. SOD2 gene expression appeared to be transcriptionally regulated by NF-κB and ZEB2, but not ZEB1. Moreover, SOD2-mediated antioxidant activity may restrict conversion of CD44L cells to CD44H cells at the early stages of EMT. These data provide novel mechanistic insights into the dynamic expression of SOD2 during EMT. In addition, we delineate a functional role for SOD2 in EMT via the influence of this antioxidant upon distinct CD44L and CD44H subsets of cancer cells that have been implicated in oral and esophageal tumor biology.


Oncogene | 2015

Manganese superoxide dismutase deficiency triggers mitochondrial uncoupling and the Warburg effect

Yong Xu; Sumitra Miriyala; Fang Fang; Vasudevan Bakthavatchalu; Teresa Noel; D M Schell; C. C. Wang; W. St. Clair; D.K. St. Clair

Manganese superoxide dismutase (MnSOD) is a mitochondrially localized primary antioxidant enzyme, known to be essential for the survival of aerobic life and to have important roles in tumorigenesis. Here, we show that MnSOD deficiency in skin tissues of MnSOD-heterozygous knockout (Sod2+/−) mice leads to increased expresson of uncoupling proteins (UCPs). When MnSOD is deficient, superoxide radical and its resulting reactive oxygen species (ROS) activate ligand binding to peroxisome proliferator-activated receptor alpha (PPARα), suggesting that the activation of PPARα signaling is a major mechanism underlying MnSOD-dependent UCPs expression that consequently triggers the PI3K/Akt/mTOR pathway, leading to increased aerobic glycolysis. Knockdown of UCPs and mTOR suppresses lactate production and increases ATP levels, suggesting that UCPs contribute to increased glycolysis. These results highlight the existence of a free radical-mediated mechanism that activates mitochondria uncoupling to reduce ROS production, which precedes the glycolytic adaptation described as the Warburg Effect.


Redox biology | 2017

A Novel Redox Regulator, MnTnBuOE-2-PyP 5+ , Enhances Normal Hematopoietic Stem/Progenitor Cell Function

Yanming Zhao; Dustin Carroll; Y. You; Luksana Chaiswing; R. Wen; Ines Batinic-Haberle; Subbarao Bondada; Ying Liang; D.K. St. Clair

The signaling of reactive oxygen species (ROS) is essential for the maintenance of normal cellular function. However, whether and how ROS regulate stem cells are unclear. Here, we demonstrate that, in transgenic mice expressing the human manganese superoxide dismutase (MnSOD) gene, a scavenger of ROS in mitochondria, the number and function of mouse hematopoietic stem/progenitor cells (HSPC) under physiological conditions are enhanced. Importantly, giving MnTnBuOE-2-PyP5+(MnP), a redox- active MnSOD mimetic, to mouse primary bone marrow cells or to C57B/L6 mice significantly enhances the number of HSPCs. Mechanistically, MnP reduces superoxide to hydrogen peroxide, which activates intracellular Nrf2 signaling leading to the induction of antioxidant enzymes, including MnSOD and catalase, and mitochondrial uncoupling protein 3. The results reveal a novel role of ROS signaling in regulating stem cell function, and suggest a possible beneficial effect of MnP in treating pathological bone marrow cell loss and in increasing stem cell population for bone marrow transplantation.


Cancer Research | 1994

Suppression of Fibrosarcoma Metastasis by Elevated Expression of Manganese Superoxide Dismutase

S.E. Safford; Terry D. Oberley; M. Urano; D.K. St. Clair


Biochemical Journal | 2000

Nuclear factor kappaB-dependent mechanisms coordinate the synergistic effect of PMA and cytokines on the induction of superoxide dismutase 2.

Kelley K. Kiningham; Yong Xu; Chotiros Daosukho; B Popova; D.K. St. Clair

Collaboration


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

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fang Fang

University of Kentucky

View shared research outputs
Top Co-Authors

Avatar

Yong Xu

University of Kentucky

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

M. Urano

University of Kentucky

View shared research outputs
Top Co-Authors

Avatar

Terry D. Oberley

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Graham W. Warren

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mary Vore

University of Kentucky

View shared research outputs
Top Co-Authors

Avatar

S. Josson

University of Kentucky

View shared research outputs
Researchain Logo
Decentralizing Knowledge