Network


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

Hotspot


Dive into the research topics where James W. Simpkins is active.

Publication


Featured researches published by James W. Simpkins.


Proceedings of the National Academy of Sciences of the United States of America | 2004

Mitochondrial localization of estrogen receptor β

Shao-Hua Yang; Ran Liu; Evelyn Perez; Yi Wen; Stanley M. Stevens; Thomas Valencia; Anne-Marie Brun-Zinkernagel; Laszlo Prokai; Yvonne Will; James A. Dykens; Peter Koulen; James W. Simpkins

Estrogen receptors (ERs) are believed to be ligand-activated transcription factors belonging to the nuclear receptor superfamily, which on ligand binding translocate into the nucleus and activate gene transcription. To date, two ERs have been identified: ERα and ERβ. ERα plays major role in the estrogen-mediated genomic actions in both reproductive and nonreproductive tissue, whereas the function of ERβ is still unclear. In this study, we used immunocytochemistry, immunoblotting, and proteomics to demonstrate that ERβ localizes to the mitochondria. In immunocytochemistry studies, ERβ was detected with two ERβ antibodies and found to colocalize almost exclusively with a mitochondrial marker in rat primary neuron, primary cardiomyocyte, and a murine hippocampal cell line. The colocalization of ERβ and mitochondrial markers was identified by both fluorescence and confocal microscopy. No translocation of ERβ into the nucleus on 17β-estradiol treatment was seen by using immunocytochemistry. Immunoblotting of purified human heart mitochondria showed an intense signal of ERβ, whereas no signals for nuclear and other organelle markers were found. Finally, purified human heart mitochondrial proteins were separated by SDS/PAGE. The 50,000–65,000 Mr band was digested with trypsin and subjected to matrix-assisted laser desorption/ionization mass spectrometric analysis, which revealed seven tryptic fragments that matched with those of ERβ. In summary, this study demonstrated that ERβ is localized to mitochondria, suggesting a role for mitochondrial ERβ in estrogen effects on this important organelle.


Neuron | 2007

Depletion of GGA3 stabilizes BACE and enhances β-secretase activity

Giuseppina Tesco; Young Ho Koh; Eugene L. Kang; Andrew N. Cameron; Shinjita Das; Miguel Sena-Esteves; Mikko Hiltunen; Shao-Hua Yang; Zhenyu Zhong; Yong Shen; James W. Simpkins; Rudolph E. Tanzi

Summary β-site APP-cleaving enzyme (BACE) is required for production of the Alzheimers disease (AD)-associated Aβ protein. BACE levels are elevated in AD brain, and increasing evidence reveals BACE as a stress-related protease that is upregulated following cerebral ischemia. However, the molecular mechanism responsible is unknown. We show that increases in BACE and β-secretase activity are due to posttranslational stabilization following caspase activation. We also found that during cerebral ischemia, levels of GGA3, an adaptor protein involved in BACE trafficking, are reduced, while BACE levels are increased. RNAi silencing of GGA3 also elevated levels of BACE and Aβ. Finally, in AD brain samples, GGA3 protein levels were significantly decreased and inversely correlated with increased levels of BACE. In summary, we have elucidated a GGA3-dependent mechanism regulating BACE levels and β-secretase activity. This mechanism may explain increased cerebral levels of BACE and Aβ following cerebral ischemia and existing in AD.


Brain Research | 2004

Increased β-secretase activity and expression in rats following transient cerebral ischemia

Yi Wen; Otuonye Onyewuchi; Shao-Hua Yang; Ran Liu; James W. Simpkins

Abstract The aberrant proteolytic processing of the amyloid precursor protein (APP) by β- and γ-secretases is key to amyloid plaque formation in Alzheimers disease (AD). Identification of an aspartyl protease as the β-secretase (β-site APP cleaving enzyme, BACE) involved in APP processing provides a pharmaceutical target for potential AD treatment. In the present studies, we demonstrate that transient cerebral ischemia in female rats caused a 30% increase in β-secretase activity. α-Secretase activity did not increase significantly. We examined protein levels of BACE1, and its analogue BACE2, in ischemic brain extracts. BACE1 protein levels increased 67%, while BACE2 protein level did not change after such a transient ischemia. Immunohistochemical studies demonstrated that BACE1 protein was increased in the ischemic neocortex, when compared with its contralateral cortex. Further, colocalization assessment indicated that BACE1 strongly associated with staining for the apoptotic marker, TUNEL. These results may partially explain epidemiological study, which demonstrate a higher incidence of dementia after stroke. Further, our results support the hypothesis that apoptosis and aberrant APP processing are correlated events in AD brain, and suggest that inhibition of BACE may have a therapeutic effect in the prevention of dementia after stroke recovery.


Proceedings of the National Academy of Sciences of the United States of America | 2003

Quinol-based cyclic antioxidant mechanism in estrogen neuroprotection

Laszlo Prokai; Katalin Prokai-Tatrai; Pál Perjési; Alevtina D. Zharikova; Evelyn Perez; Ran Liu; James W. Simpkins

Substantial evidence now exists that intrinsic free-radical scavenging contributes to the receptor-independent neuroprotective effects of estrogens. This activity is inherently associated with the presence of a phenolic A-ring in the steroid. We report a previously unrecognized antioxidant cycle that maintains the “chemical shield” raised by estrogens against the most harmful reactive oxygen species, the hydroxyl radical (•OH) produced by the Fenton reaction. In this cycle, the capture of •OH was shown to produce a nonphenolic quinol with no affinity to the estrogen receptors. This quinol is then rapidly converted back to the parent estrogen via an enzyme-catalyzed reduction by using NAD(P)H as a coenzyme (reductant) and, unlike redox cycling of catechol estrogens, without the production of reactive oxygen species. Due to this process, protection of neuronal cells against oxidative stress is also possible by quinols that essentially act as prodrugs for the active hormone. We have shown that the quinol obtained from a 17β-estradiol derivative was, indeed, able to attenuate glutamate-induced oxidative stress in cultured hippocampus-derived HT-22 cells. Estrone quinol was also equipotent with its parent estrogen in reducing lesion volume in ovariectomized rats after transient middle carotid artery occlusion followed by a 24-h reperfusion. These findings may establish the foundation for a rational design of neuroprotective antioxidants focusing on steroidal quinols as unique molecular leads.


Journal of Neurochemistry | 2002

Modulation of Aβ peptides by estrogen in mouse models

Hui Zheng; H. Xu; S. N. Uljon; R. Gross; K. Hardy; J. Gaynor; J. Lafrancois; James W. Simpkins; L. M. Refolo; Suzana S. Petanceska; Rong Wang; Karen Duff

Clinical studies have shown that estrogen deprivation through menopause is a risk factor in both the initiation and progression of Alzheimers disease (AD) and that estrogen replacement therapy may be protective. One of the major pathological features in the human AD brain is the senile plaque, a proteinaceous structure composed mainly of heterogeneous peptides collectively known as A‐beta (Aβ). In vitro studies have linked estrogen with Aβ modulation, suggesting that one‐way that estrogen depletion at menopause may exacerbate the features of AD is through Aβ accumulation. To test this, two studies were performed on transgenic models of amyloidosis. Firstly, transgenic mice without detectable amyloid aggregates were subjected to ovariectomy and estradiol supplementation, and Aβ levels were assessed. Secondly, the effects of estrogen modulation were assessed in mice at an age when plaques would be forming initially. Overall, Aβ levels were higher in estrogen‐deprived mice than intact mice, and this effect could be reversed through the administration of estradiol. These data suggest that, in vivo, estrogen depletion leads to the accumulation of Aβ in the CNS, which can be reversed through replacement of estradiol. These results provide evidence that post‐menopausal estrogen depletion may be linked to an increased risk of AD through Aβ modulation.


Journal of Biological Chemistry | 2011

Alternative Mitochondrial Electron Transfer as a Novel Strategy for Neuroprotection

Yi Wen; Wenjun Li; Ethan Poteet; Luokun Xie; Cong Tan; Liang-Jun Yan; Xiaohua Ju; Ran Liu; Hai Qian; Marian Marvin; Matthew S. Goldberg; Hua She; Zixu Mao; James W. Simpkins; Shao-Hua Yang

Neuroprotective strategies, including free radical scavengers, ion channel modulators, and anti-inflammatory agents, have been extensively explored in the last 2 decades for the treatment of neurological diseases. Unfortunately, none of the neuroprotectants has been proved effective in clinical trails. In the current study, we demonstrated that methylene blue (MB) functions as an alternative electron carrier, which accepts electrons from NADH and transfers them to cytochrome c and bypasses complex I/III blockage. A de novo synthesized MB derivative, with the redox center disabled by N-acetylation, had no effect on mitochondrial complex activities. MB increases cellular oxygen consumption rates and reduces anaerobic glycolysis in cultured neuronal cells. MB is protective against various insults in vitro at low nanomolar concentrations. Our data indicate that MB has a unique mechanism and is fundamentally different from traditional antioxidants. We examined the effects of MB in two animal models of neurological diseases. MB dramatically attenuates behavioral, neurochemical, and neuropathological impairment in a Parkinson disease model. Rotenone caused severe dopamine depletion in the striatum, which was almost completely rescued by MB. MB rescued the effects of rotenone on mitochondrial complex I-III inhibition and free radical overproduction. Rotenone induced a severe loss of nigral dopaminergic neurons, which was dramatically attenuated by MB. In addition, MB significantly reduced cerebral ischemia reperfusion damage in a transient focal cerebral ischemia model. The present study indicates that rerouting mitochondrial electron transfer by MB or similar molecules provides a novel strategy for neuroprotection against both chronic and acute neurological diseases involving mitochondrial dysfunction.


Brain Research | 2004

Estrogen attenuates nuclear factor-kappa B activation induced by transient cerebral ischemia

Yi Wen; Shao-Hua Yang; Ran Liu; Evelyn Perez; Kun Don Yi; Peter Koulen; James W. Simpkins

The protective effects of estrogens have been widely reported in a number of animal and cell culture models, but the molecular mechanisms of this potent neuroprotective activity are not well understood. Both in vitro and in vivo studies indicate that in the central nervous system and peripheral tissues, estrogen treatment reduces cytokine production and inflammatory responses. Nuclear factor-kappa B (NFkappaB) plays an essential role in the regulation of post-ischemic inflammation, which is detrimental to recovery from an ischemic stroke. We investigated the role of NFkappaB in neuronal survival in rats that received transient middle cerebral artery (MCA) occlusion, and observed that this transient cerebral ischemia induced substantial apoptosis and inflammatory responses, including IkappaB phosphorylation, NF-kappaB activation and iNOS over-expression. 17 beta-estradiol (E2) treatment produced strong protective effects by reducing infarct volume, neuronal apoptosis, and inflammatory responses. These findings provide evidence for a novel molecular and cellular interaction between the sex hormone and the immunoresponsive system. These studies also provide evidence that suppression of post-ischemic inflammation may play a critical role in estrogen-mediated neuroprotection.


Biochimica et Biophysica Acta | 2010

Mitochondrial mechanisms of estrogen neuroprotection.

James W. Simpkins; Kun Don Yi; Shao-Hua Yang; James A. Dykens

Mitochondria have become a primary focus in our search not only for the mechanism(s) of neuronal death but also for neuroprotective drugs and therapies that can delay or prevent Alzheimers disease and other chronic neurodegenerative conditions. This is because mitochrondria play a central role in regulating viability and death of neurons, and mitochondrial dysfunction has been shown to contribute to neuronal death seen in neurodegenerative diseases. In this article, we review the evidence for the role of mitochondria in cell death and neurodegeneration and provide evidence that estrogens have multiple effects on mitochondria that enhance or preserve mitochondrial function during pathologic circumstances such as excitotoxicity, oxidative stress, and others. As such, estrogens and novel non-hormonal analogs have come to figure prominently in our efforts to protect neurons against both acute brain injury and chronic neurodegeneration.


Aging Cell | 2014

Acarbose, 17-α-estradiol, and nordihydroguaiaretic acid extend mouse lifespan preferentially in males

David E. Harrison; Randy Strong; David B. Allison; Bruce N. Ames; Clinton M. Astle; Hani Atamna; Elizabeth Fernandez; Kevin Flurkey; Martin A. Javors; Nancy L. Nadon; James F. Nelson; Scott D. Pletcher; James W. Simpkins; Daniel Smith; J. Erby Wilkinson; Richard A. Miller

Four agents — acarbose (ACA), 17‐α‐estradiol (EST), nordihydroguaiaretic acid (NDGA), and methylene blue (MB) — were evaluated for lifespan effects in genetically heterogeneous mice tested at three sites. Acarbose increased male median lifespan by 22% (P < 0.0001), but increased female median lifespan by only 5% (P = 0.01). This sexual dimorphism in ACA lifespan effect could not be explained by differences in effects on weight. Maximum lifespan (90th percentile) increased 11% (P < 0.001) in males and 9% (P = 0.001) in females. EST increased male median lifespan by 12% (P = 0.002), but did not lead to a significant effect on maximum lifespan. The benefits of EST were much stronger at one test site than at the other two and were not explained by effects on body weight. EST did not alter female lifespan. NDGA increased male median lifespan by 8–10% at three different doses, with P‐values ranging from 0.04 to 0.005. Females did not show a lifespan benefit from NDGA, even at a dose that produced blood levels similar to those in males, which did show a strong lifespan benefit. MB did not alter median lifespan of males or females, but did produce a small, statistically significant (6%, P = 0.004) increase in female maximum lifespan. These results provide new pharmacological models for exploring processes that regulate the timing of aging and late‐life diseases, and in particular for testing hypotheses about sexual dimorphism in aging and health.


Journal of Biological Chemistry | 2004

Transient Cerebral Ischemia Induces Aberrant Neuronal Cell Cycle Re-entry and Alzheimer's Disease-like Tauopathy in Female Rats

Yi Wen; Shao-Hua Yang; Ran Liu; Anne Marie Brun-Zinkernagel; Peter Koulen; James W. Simpkins

Aberrant mitosis occurs in many tauopathy-related neurodegenerative diseases and is believed to precede the formation of neurofibrillary tangles. In this study, we report for the first time that transient cerebral ischemia induces aberrant mitotic proteins and hyperphosphorylation of tau protein with neurofibrillary tangle-like conformational epitopes in adult female rat cortex. Following transient cerebral ischemia in rats, initiation of apoptosis precedes and is potentially integrated with subsequent aberrant mitosis and tau hyperphosphorylation. Furthermore, inhibition of mitosis-related cyclin-dependent kinases (Cdks) by roscovitine significantly reduced the hyperphosphorylation of tau. Administration of the female sex steroid and potent neuroprotective agent, 17β-estradiol, reduced ischemia-reperfusion-induced cerebral damage and the subsequent aberrant mitosis and tauopathies. These results provide a neuropathological basis for the higher prevalence of dementia in stroke patients and support the hypothesis that apoptosis and aberrant mitosis are integrated pathological events in neurons that may play a critical role in the development of Alzheimers disease and other tauopathy-related neuropathology.

Collaboration


Dive into the James W. Simpkins's collaboration.

Top Co-Authors

Avatar

Shao-Hua Yang

University of North Texas Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Yi Wen

University of North Texas Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Ran Liu

University of North Texas Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Evelyn Perez

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Marianna E. Jung

University of North Texas Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Jane G. Wigginton

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Joshua W. Gatson

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Xuefang Ren

West Virginia University

View shared research outputs
Top Co-Authors

Avatar

Douglas F. Covey

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Laszlo Prokai

University of North Texas Health Science Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge