Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters











Publication year range
1.
Adv Exp Med Biol ; 1232: 409-414, 2020.
Article in English | MEDLINE | ID: mdl-31893438

ABSTRACT

Nakamura et al. examined the evidence, using a discovery and a validation database, that amyloid-ß precursor protein (APP)669-711/amyloid-ß (Aß)1-42 and Aß1-40/Aß1-42 ratios, and composites based on traditional statistics; they concluded that these may be useful as biomarkers of Alzheimer's Disease (AD). We reexamined the same datasets, each of which included cognitively normal individuals (CN), individuals with mild cognitive impairment (MCI) and individuals with AD. We used fractal self-similar analyses and reexamined their data from (1) the Japanese National Center for Geriatrics and Gerontology (NCGG) (discovery database) and (2) the Australian Imaging, Biomarker and Lifestyle Study of Ageing (AIBL) cohort (validation database). Results: Using our methods, the three groups of individuals were found to be self-similar, i.e., they could not be differentiated quantitatively, in contrast to the findings of Nakamura et al. Conclusion: Appropriate biomarkers need further study. Our results suggest that APP669-711/Aß1-42 and Aß1-40/Aß1-42 ratios and their composites may not be valid biomarkers of AD, when reexamined using fractal methods for comparing biomarkers across populations.


Subject(s)
Alzheimer Disease , Biomarkers , Alzheimer Disease/diagnosis , Amyloid beta-Peptides , Humans
2.
Adv Exp Med Biol ; 1072: 281-285, 2018.
Article in English | MEDLINE | ID: mdl-30178359

ABSTRACT

Luongo et al. found that the mitochondrial Na+/Ca2+ exchanger (NCLX) was essential for Ca2+ homeostasis and viability. Here, we re-analyze their data in terms of fractal self-similarity and quantitative difference (QD). We calculated the 7-dimension data from NCLX conditional loss-of-function mouse models, and the 9-dimension data from NCLX overexpression (NCLX-Tg) models. RESULTS: The 9-dimension data of the NCLX-Tg and its tTA control were partially self-similar to each other, while the 7-dimension data in NCLX knockout models were not. CONCLUSION: The NCLX may be necessary but is not sufficient for Ca2+ homeostasis and viability.


Subject(s)
Calcium/metabolism , Cell Survival/physiology , Homeostasis/physiology , Models, Theoretical , Sodium-Calcium Exchanger/metabolism , Algorithms , Animals , Calcium Signaling/physiology , Mice , Mitochondria/metabolism
3.
Adv Exp Med Biol ; 977: 419-424, 2017.
Article in English | MEDLINE | ID: mdl-28685473

ABSTRACT

Exercise is essential in regulating energy metabolism. Exercise activates cellular, molecular, and biochemical pathways with regulatory roles in training response adaptation. Among them, endurance/strength training of an individual has been shown to activate its respective signal transduction pathways in skeletal muscle. This was further studied from the viewpoint of quantitative difference (QD). For the mean values, [Formula: see text], of two sets of data, their QD is defined as [Formula: see text] ([Formula: see text]). The function-specific homeostasis (FSH) of a function of a biosystem is a negative-feedback response of the biosystem to maintain the function-specific conditions inside the biosystem so that the function is perfectly performed. A function in/far from its FSH is called a normal/dysfunctional function. A cellular normal function can resist the activation of other signal transduction pathways so that there are normal function-specific signal transduction pathways which full activation maintains the normal function. RESULTS: An acute endurance/strength training may be dysfunctional, but its regular training may be normal. The normal endurance/strength training of an individual may resist the activation of other signal transduction pathways in skeletal muscle so that there may be normal endurance/strength training-specific signal transduction pathways (NEPs/NSPs) in skeletal muscle. The endurance/strength training may activate NSPs/NEPs, but the QD from the control is smaller than 0.80. The simultaneous activation of both NSPs and NEPs may enhance their respective activation, and the QD from the control is larger than 0.80. The low level laser irradiation pretreatment of rats may promote the activation of NSPs in endurance training skeletal muscle. CONCLUSION: There may be NEPs/NSPs in skeletal muscle trained by normal endurance/strength training.


Subject(s)
Exercise/physiology , Physical Endurance/physiology , Signal Transduction , Adaptation, Physiological/physiology , Energy Metabolism/physiology , Humans , Muscle Strength/physiology , Muscle, Skeletal/physiology , Resistance Training
4.
Sci Rep ; 6: 37033, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27845401

ABSTRACT

Post-ischemic activation of NMDA receptors (NMDARs) has been linked to NMDAR subunit-specific signaling that mediates pro-survival or pro-death activity. Although extensive studies have been performed to characterize the role of GluN2A and GluN2B following ischemia, there is less understanding regarding the regulation of GluN2C. Here, we show that GluN2C expression is increased in acute hippocampal slices in response to ischemia. Strikingly, GluN2C knockout mice, following global cerebral ischemia, exhibit greater neuronal death in the CA1 area of the hippocampus and reduced spatial working memory compared to wild-type mice. Moreover, we find that GluN2C-expressing hippocampal neurons show marked resistance to NMDA-induced toxicity and reduced calcium influx. Using both in vivo and in vitro experimental models of ischemia, we demonstrate a neuroprotective role of GluN2C, suggesting a mechanism by which GluN2C is upregulated to promote neuronal survival following ischemia. These results may provide insights into development of NMDAR subunit-specific therapeutic strategies to protect neurons from excitotoxicity.


Subject(s)
Brain Ischemia/metabolism , Calcium Signaling , Gene Expression Regulation , Hippocampus/metabolism , Neurons/metabolism , Neuroprotection , Receptors, N-Methyl-D-Aspartate/biosynthesis , Animals , Brain Ischemia/genetics , Brain Ischemia/pathology , Cell Death , Hippocampus/pathology , Male , Mice , Mice, Knockout , Neurons/pathology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/genetics
5.
Hippocampus ; 25(1): 62-71, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25139533

ABSTRACT

Oligodendrocytes are the predominant cell type in white matter and are highly vulnerable to ischemic injury. The role of oligodendrocyte dysfunction in ischemic brain injury is unknown. In this study, we used a 24-amino acid peptide S14G-Humanin (HNG) to examine oligodendrogenesis and neurological functional recovery in a hypoxic/ischemic (H/I) neonatal model. Intraperitoneal HNG pre-treatment decreased infarct volume following H/I injury. Delayed HNG treatment 24 h after H/I injury did not reduce infarct volume but did decrease neurological deficits and brain atrophy. Delayed HNG treatment did not attenuate axonal demyelination at 48 h after H/I injury. However, at 14 d after H/I injury, delayed HNG treatment increased axonal remyelination, the thickness of corpus callosum at the midline, the number of Olig2(+) /BrdU(+) cells, and levels of brain-derived neurotrophic factor (BDNF). Our results suggest that targeting oligodendrogenesis via delayed HNG treatment may represent a promising approach for the treatment of stroke.


Subject(s)
Hypoxia-Ischemia, Brain/drug therapy , Intracellular Signaling Peptides and Proteins/pharmacology , Neurogenesis/drug effects , Oligodendroglia/drug effects , Recovery of Function/drug effects , Animals , Animals, Newborn , Atrophy/pathology , Axons/drug effects , Axons/pathology , Brain-Derived Neurotrophic Factor/drug effects , Cerebral Infarction/drug therapy , Cerebral Infarction/pathology , Cerebral Infarction/physiopathology , Disease Models, Animal , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/physiopathology , Intracellular Signaling Peptides and Proteins/administration & dosage , Random Allocation , Rats , Rats, Sprague-Dawley
6.
Mol Cell Endocrinol ; 389(1-2): 84-91, 2014 May 25.
Article in English | MEDLINE | ID: mdl-24508637

ABSTRACT

17ß-estradiol (E2) has been implicated to play a critical role in neuroprotection, synaptic plasticity, and cognitive function. Classically, the role of gonadal-derived E2 in these events is well established, but the role of brain-derived E2 is less clear. To address this issue, we investigated the expression, localization, and modulation of aromatase and local E2 levels in the hippocampus following global cerebral ischemia (GCI) in adult ovariectomized rats. Immunohistochemistry (IHC) revealed that the hippocampal regions CA1, CA3 and dentate gyrus (DG) exhibited high levels of immunoreactive aromatase staining, with aromatase being co-localized primarily in neurons in non-ischemic animals. Following GCI, aromatase became highly expressed in GFAP-positive astrocytes in the hippocampal CA1 region at 2-3 days post GCI reperfusion. An ELISA for E2 and IHC for E2 confirmed the GCI-induced elevation of local E2 in the CA1 region and that the increase in local E2 occurred in astrocytes. Furthermore, central administration of aromatase antisense (AS) oligonucleotides, but not missense (MS) oligonucleotides, blocked the increase in aromatase and local E2 in astrocytes after GCI, and resulted in a significant increase in GCI-induced hippocampal CA1 region neuronal cell death and neuroinflammation. As a whole, these results suggest that brain-derived E2 exerts important neuroprotective and anti-inflammatory actions in the hippocampal CA1 region following GCI.


Subject(s)
Anti-Inflammatory Agents/metabolism , Dentate Gyrus/metabolism , Estrogens/metabolism , Neuroprotective Agents/metabolism , Animals , Aromatase/metabolism , Astrocytes/metabolism , Brain Ischemia/metabolism , CA1 Region, Hippocampal/metabolism , Cell Death/physiology , Estradiol/metabolism , Female , Inflammation/metabolism , Neurons/metabolism , Rats , Rats, Sprague-Dawley
7.
Mol Cell Endocrinol ; 389(1-2): 2-6, 2014 May 25.
Article in English | MEDLINE | ID: mdl-24462786

ABSTRACT

Since basic scientific studies in the 1990s revealed dramatic gender differences in neurological damage from cerebral ischemia, significant evidence has accumulated for a neuroprotective role of ovarian-derived 17ß-Estradiol (E2). Intriguingly, observational studies have further suggested that early and prolonged loss of ovarian E2 (premature menopause) leads to a doubled lifetime risk for dementia and a fivefold increased risk of mortality from neurological disorders, but some controversy remains. Here, we briefly summarize and analyze clinical cohort studies assessing the detrimental neurological outcomes of premature menopause. Furthermore, we discuss current basic science studies elucidating the molecular mechanisms underlying the enhanced risk of neurological disease in prematurely menopausal women and the "window of opportunity" for estrogen benefit. Finally, we highlight four critical issues in the field that require collaboration between basic scientists and clinicians for successful resolution, with the ultimate goal of maintaining optimal neurological health in prematurely menopausal women.


Subject(s)
Menopause, Premature/physiology , Nervous System Diseases/physiopathology , Cohort Studies , Estrogens/metabolism , Female , Humans , Menopause, Premature/metabolism , Nervous System Diseases/metabolism , Risk , Women's Health
8.
Brain ; 136(Pt 5): 1432-45, 2013 May.
Article in English | MEDLINE | ID: mdl-23474850

ABSTRACT

Females who enter menopause prematurely via bilateral ovariectomy (surgical menopause) have a significantly increased risk for cognitive decline and dementia. To help elucidate the mechanisms underlying this phenomenon, we used an animal model of surgical menopause, long-term (10-week) bilateral ovariectomy in female rats. Herein, we demonstrate that long-term oestrogen deprivation dramatically increases sensitivity of the normally resistant hippocampal CA3 region to ischaemic stress, an effect that was gender-specific, as it was not observed in long-term orchiectomized males. Furthermore, the enhanced damage to the CA3 region correlated with a worse cognitive outcome after ischaemic stress. Long-term ovariectomized rats also displayed a robust hyperinduction of Alzheimer's disease-related proteins in the CA3 region and a switch in amyloid precursor protein processing from non-amyloidogenic to amyloidogenic following ischaemic stress CA3 hypersensitivity also extended to an Alzheimer's disease-relevant insult, as the CA3 region of long-term ovariectomized rats was profoundly hypersensitive to the neurotoxic effects of amyloid-ß1-42, the most amyloidogenic form of the amyloid-ß peptide. Additional studies revealed that CA3 region hypersensitivity, Alzheimer's disease-related protein induction, and amyloidogenesis are mediated by a NADPH oxidase/superoxide/c-Jun N-terminal kinase/c-Jun signalling pathway, involving both transcriptional and post-translational mechanisms. In addition, while 17ß-oestradiol replacement at the end of the long-term oestrogen deprivation period could not prevent CA3 hypersensitivity and amyloidogenesis, if 17ß-oestradiol was initiated at the time of ovariectomy and maintained throughout the 10-week oestrogen deprivation period, it completely prevented these events, providing support for the 'critical window' hypothesis for oestrogen replacement therapy benefit. Collectively, these findings may help explain the increased risk of cognitive decline and dementia observed in women following surgical menopause, and they provide increased support that early 17ß-oestradiol replacement is critical in preventing the negative neural effects associated with bilateral ovariectomy.


Subject(s)
Amyloid beta-Peptides/biosynthesis , Amyloid beta-Peptides/toxicity , CA3 Region, Hippocampal/metabolism , Menopause/metabolism , Nerve Degeneration/metabolism , Ovariectomy , Peptide Fragments/biosynthesis , Peptide Fragments/toxicity , Stress, Physiological/physiology , Amyloid beta-Peptides/metabolism , Animals , CA3 Region, Hippocampal/pathology , Female , Male , Models, Animal , Nerve Degeneration/pathology , Ovariectomy/adverse effects , Peptide Fragments/metabolism , Rats , Rats, Sprague-Dawley
9.
Steroids ; 78(6): 614-23, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23276632

ABSTRACT

The steroid hormone, 17ß-estradiol (E2) has been reported to enhance executive functions that are known to be mediated by the prefrontal cortex (PFC), although the underlying mechanisms remain unclear. To shed light on the potential mechanisms, we examined the effect of E2 in vivo upon spine density in the rat PFC and the somatosensory cortex (SSC), which has been implicated to be a transient storage site for information that can also contribute to working memory. The results revealed that E2 significantly enhanced the number of dendritic spines in both the SSC and PFC, as well as the expression of spinophilin. In vitro studies revealed further mechanistic insights by demonstrating that E2 enhanced AMPA GluR1 receptor expression and excitatory glutamatergic synapse formation in rat cortical neurons, without an effect upon inhibitory GABAergic synapse formation. Furthermore, E2 rapidly enhanced ERK and Akt activation in cortical neurons, and inhibitors of ERK and Akt activation significantly attenuated E2 induction of excitatory glutamatergic synapses. Administration of E2-BSA likewise significantly enhanced excitatory glutamatergic synapses in cortical neurons, and administration of an ER antagonist, ICI182,780 and a non-NMDA receptor antagonist (NBQX) significantly attenuated the effect of E2 upon enhancement of excitatory glutamatergic synapses, suggesting mediation by extranuclear estrogen receptors and involvement of non-NMDA receptor activation and signaling. As a whole, the studies demonstrate that E2 enhances spine density in both the PFC and SSC, and that E2 enhances excitatory glutamatergic synapse formation in cortical neurons via a rapid extranuclear ER-mediated signaling mechanism that involves up-regulation of GluR1 and mediation by Akt and ERK signaling pathways.


Subject(s)
Dendritic Spines/drug effects , Estradiol/pharmacology , Prefrontal Cortex/drug effects , Somatosensory Cortex/drug effects , Synapses/drug effects , Animals , Dendritic Spines/metabolism , Estradiol/metabolism , Female , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Somatosensory Cortex/metabolism , Synapses/metabolism
10.
Steroids ; 78(6): 624-32, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23178162

ABSTRACT

Surgically menopausal women incur a 2- to 5-fold increased risk for dementia and mortality from neurological diseases, but the mechanisms underlying these increased risks remain unclear. Previously, we demonstrated that after global cerebral ischemia (GCI), 17ß-estradiol (E2 or estrogen) suppresses hippocampal elevation of the Wnt antagonist Dickkopf-1 (Dkk1), a neurodegenerative factor. We, thus, hypothesized that prolonged loss of E2 may lead to dysregulation of neural Dkk1 and Wnt/ß-Catenin signaling, which could contribute to an increased risk of neurodegeneration. To test this hypothesis, we examined the effect of short-term (1 week - STED) and long-term E2 deprivation (10 weeks - LTED) via ovariectomy upon basal and E2-regulated Dkk1 levels and Wnt/ß-Catenin signaling in the hippocampal CA1 region following GCI. In STED rats, E2 exerted robust neuroprotection against GCI, suppressed post-ischemic elevation of Dkk1, and enhanced pro-survival Wnt/ß-Catenin signaling, effects that were lost in LTED rats. Intriguingly, LTED rats displayed modest basal changes in Dkk1 and survivin expression. Further work showed that c-Jun N-terminal Kinase (JNK) mediated GCI-induced changes in Dkk1 and survivin, and JNK inhibition afforded neuroprotection in LTED rats. Finally, we extended our findings to natural aging, as 24-month-old, reproductively senescent female rats also displayed a modest increase in basal Dkk1 in the CA1, which consistently co-localized with the apoptotic marker TUNEL after GCI and coincided with a loss of E2 neuroprotection. As a whole, this study supports the "critical period hypothesis" and further suggests that perimenopausal estradiol replacement may prevent neurodegenerative changes in the hippocampus by maintaining favorable Wnt/ß-Catenin signaling.


Subject(s)
CA1 Region, Hippocampal/cytology , Estradiol/deficiency , Neurons/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Estradiol/pharmacology , Female , Neurons/cytology , Neurons/drug effects , Rats , Rats, Sprague-Dawley
11.
Oncotarget ; 4(1): 18-28, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23248157

ABSTRACT

Glioma development is a multistep process, involving alterations in genetic and epigenetic mechanisms. Understanding the mechanisms and enzymes that promote epigenetic changes in gliomas are urgently needed to identify novel therapeutic targets. We examined the role of histone demethylase KDM1 in glioma progression. KDM1 was overexpressed in gliomas and its expression positively correlated with histological malignancy. Knockdown of KDM1 expression or its pharmacological inhibition using pargyline or NCL-1 significantly reduced the proliferation of glioma cells. Inhibition of KDM1 promoted up regulation of the p53 target genes p21 and PUMA. Patient-derived primary GBM cells expressed high levels of KDM1 and pharmacological inhibition of KDM1 decreased their proliferation. Further, KDM1 inhibition reduced the expression of stemness markers CD133 and nestin in GBM cells. Mouse xenograft assays revealed that inhibition of KDM1 significantly reduced glioma xenograft tumor growth. Inhibition of KDM1 increased levels of H3K4-me2 and H3K9-Ac histone modifications, reduced H3K9-me2 modification and promoted expression of p53 target genes (p21 and PUMA), leading to apoptosis of glioma xenograft tumors. Our results suggest that KDM1 is overexpressed in gliomas and could be a potential therapeutic target for the treatment of gliomas.


Subject(s)
Brain Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Glioma/drug therapy , Histone Demethylases/antagonists & inhibitors , Xenograft Model Antitumor Assays , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Benzamides/pharmacology , Benzamides/therapeutic use , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclopropanes/pharmacology , Cyclopropanes/therapeutic use , Enzyme Inhibitors/therapeutic use , Female , Gene Expression Regulation, Neoplastic/drug effects , Glioma/metabolism , Glioma/pathology , Histone Demethylases/genetics , Histone Demethylases/metabolism , Humans , Immunohistochemistry , MCF-7 Cells , Mice , Mice, Inbred NOD , Mice, SCID , Pargyline/pharmacology , Pargyline/therapeutic use , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
12.
PLoS One ; 7(4): e34504, 2012.
Article in English | MEDLINE | ID: mdl-22485176

ABSTRACT

BACKGROUND: Oxidative stress is known to play an important role in the pathology of traumatic brain injury. Mitochondria are thought to be the major source of the damaging reactive oxygen species (ROS) following TBI. However, recent work has revealed that the membrane, via the enzyme NADPH oxidase can also generate the superoxide radical (O(2)(-)), and thereby potentially contribute to the oxidative stress following TBI. The current study thus addressed the potential role of NADPH oxidase in TBI. METHODOLOGY/PRINCIPAL FINDINGS: The results revealed that NADPH oxidase activity in the cerebral cortex and hippocampal CA1 region increases rapidly following controlled cortical impact in male mice, with an early peak at 1 h, followed by a secondary peak from 24-96 h after TBI. In situ localization using oxidized hydroethidine and the neuronal marker, NeuN, revealed that the O(2)(-) induction occurred in neurons at 1 h after TBI. Pre- or post-treatment with the NADPH oxidase inhibitor, apocynin markedly inhibited microglial activation and oxidative stress damage. Apocynin also attenuated TBI-induction of the Alzheimer's disease proteins ß-amyloid and amyloid precursor protein. Finally, both pre- and post-treatment of apocynin was also shown to induce significant neuroprotection against TBI. In addition, a NOX2-specific inhibitor, gp91ds-tat was also shown to exert neuroprotection against TBI. CONCLUSIONS/SIGNIFICANCE: As a whole, the study demonstrates that NADPH oxidase activity and superoxide production exhibit a biphasic elevation in the hippocampus and cortex following TBI, which contributes significantly to the pathology of TBI via mediation of oxidative stress damage, microglial activation, and AD protein induction in the brain following TBI.


Subject(s)
Brain Injuries/enzymology , Membrane Glycoproteins/metabolism , Microglia/physiology , NADPH Oxidases/metabolism , Neurons/enzymology , Acetophenones/pharmacology , Amyloid beta-Peptides/metabolism , Animals , Brain Edema/enzymology , Brain Edema/pathology , Brain Injuries/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Enzyme Activation , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Male , Membrane Glycoproteins/antagonists & inhibitors , Mice , Microglia/drug effects , Microglia/metabolism , NADPH Oxidase 2 , NADPH Oxidases/antagonists & inhibitors , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Oxidation-Reduction , Oxidative Stress , Superoxides/metabolism
13.
Mol Cancer Ther ; 11(5): 1174-82, 2012 May.
Article in English | MEDLINE | ID: mdl-22442308

ABSTRACT

Gliomas are the most common and devastating central nervous system neoplasms. A gender bias exists in their development: females are at lower risk than males, implicating estrogen-mediated protective effects. Estrogen functions are mediated by two estrogen receptor (ER) subtypes: ERα, which functions as tumor promoter, and ERß, which functions as tumor suppressor. We examined the potential use of ERß agonists as a novel therapeutic to curb the growth of gliomas. Western analysis of six glioma model cells showed detectable expression of ERß with little or no ERα. Treatment of glioma cells with ERß agonists resulted in significant decrease in proliferation. Immunohistochemical analysis of tumor tissues revealed that ERß expression is downregulated in high-grade gliomas. We found that ERß agonists promote both expression and tumor-suppressive functions of ERß in glioma cells. Liquiritigenin, a plant-derived ERß agonist significantly reduced in vivo tumor growth in a xenograft model. Compared with control mice, animals treated with liquiritigenin had greater than 50% reduction in tumor volume and size. Immunohistochemical analysis of tumors revealed a significant increase in the nuclear ERß expression with a concomitant decrease in cell proliferation in the liquiritigenin-treated group. Our results suggest that ERß signaling has a tumor-suppressive function in gliomas. Because ERß agonists are currently in clinical trials and are well tolerated with fewer side effects, identification of an ERß agonist as a therapeutic agent can be readily extended to clinical use with current chemotherapies, providing an additional tool for enhancing survival in glioma patients.


Subject(s)
Antineoplastic Agents/pharmacology , Estrogen Receptor beta/agonists , Glioma/drug therapy , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Estrogen Receptor beta/genetics , Female , Flavanones/pharmacology , Gene Expression/drug effects , Glioma/genetics , Glioma/pathology , Humans , Mice , Mice, Nude , Protein Transport , Signal Transduction
14.
Front Neuroendocrinol ; 33(1): 85-104, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22079780

ABSTRACT

17ß-Estradiol (estradiol or E2) is implicated as a neuroprotective factor in a variety of neurodegenerative disorders. This review focuses on the mechanisms underlying E2 neuroprotection in cerebral ischemia, as well as emerging evidence from basic science and clinical studies, which suggests that there is a "critical period" for estradiol's beneficial effect in the brain. Potential mechanisms underlying the critical period are discussed, as are the neurological consequences of long-term E2 deprivation (LTED) in animals and in humans after natural menopause or surgical menopause. We also summarize the major clinical trials concerning postmenopausal hormone therapy (HT), comparing their outcomes with respect to cardiovascular and neurological disease and discussing their relevance to the critical period hypothesis. Finally, potential caveats, controversies and future directions for the field are highlighted and discussed throughout the review.


Subject(s)
Brain Ischemia/prevention & control , Estradiol/therapeutic use , Neurodegenerative Diseases/prevention & control , Neuroprotective Agents/therapeutic use , Animals , Critical Period, Psychological , Estrogen Receptor alpha/physiology , Estrogen Replacement Therapy , Female , Humans , Menopause , Mice , Oxidative Stress/drug effects , Rats , Receptors, Estrogen , Receptors, G-Protein-Coupled/physiology
15.
J Cell Biol ; 195(5): 765-79, 2011 Nov 28.
Article in English | MEDLINE | ID: mdl-22105352

ABSTRACT

VPS35, a major component of the retromer complex, is important for endosome-to-Golgi retrieval of membrane proteins. Although implicated in Alzheimer's disease (AD), how VPS35 regulates AD-associated pathology is unknown. In this paper, we show that hemizygous deletion of Vps35 in the Tg2576 mouse model of AD led to earlier-onset AD-like phenotypes, including cognitive memory deficits, defective long-term potentiation, and impaired postsynaptic glutamatergic neurotransmission in young adult age. These deficits correlated well with an increase of ß-amyloid peptide (Aß) level in the mutant hippocampus. We further demonstrate that VPS35 is predominantly expressed in pyramidal neurons of young adult hippocampus and interacts with BACE1, a protease responsible for Aß production. Loss of VPS35 function in the mouse hippocampus increased BACE1 activity. Suppression of VPS35 expression in culture decreased BACE1 trans-Golgi localization but enriched it in endosomes. These results demonstrate an essential role for VPS35 in suppression of AD neuropathology and in inhibition of BACE1 activation and Aß production by promoting BACE1 endosome-to-Golgi retrieval.


Subject(s)
Alzheimer Disease/genetics , Haploinsufficiency , Vesicular Transport Proteins/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Cerebral Cortex/metabolism , Endosomes/metabolism , HEK293 Cells , Hippocampus/metabolism , Humans , Mice , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/physiology , trans-Golgi Network/metabolism
16.
PLoS One ; 6(10): e27039, 2011.
Article in English | MEDLINE | ID: mdl-22046440

ABSTRACT

BACKGROUND: Recent studies demonstrate that acetylation of the transcription factor, p53 on lysine(373) leads to its enhanced stabilization/activity and increased susceptibility of cells to stress. However, it is not known whether acetylation of p53 is altered in the hippocampus following global cerebral ischemia (GCI) or is regulated by the hormone, 17ß-estradiol (17ß-E(2)), and thus, this study examined these issues. METHODOLOGY/PRINCIPAL FINDINGS: The study revealed that Acetyl p53-Lysine(373) levels were markedly increased in the hippocampal CA1 region after GCI at 3 h, 6 h and 24 h after reperfusion, an effect strongly attenuated by 17ß-E(2). 17ß-E(2) also enhanced interaction of p53 with the ubiquitin ligase, Mdm2, increased ubiquitination of p53, and induced its down-regulation, as well as attenuated elevation of the p53 transcriptional target, Puma. We also observed enhanced acetylation of p53 at a different lysine (Lys(382)) at 3 h after reperfusion, and 17ß-E(2) also markedly attenuated this effect. Furthermore, administration of an inhibitor of CBP/p300 acetyltransferase, which acetylates p53, was strongly neuroprotective of the CA1 region following GCI. In long-term estrogen deprived (LTED) animals, the ability of 17ß-E(2) to attenuate p53 acetylation was lost, and intriguingly, Acetyl p53-Lysine(373) levels were markedly elevated in sham (non-ischemic) LTED animals. Finally, intracerebroventricular injections of Gp91ds-Tat, a specific NADPH oxidase (NOX2) inhibitor, but not the scrambled tat peptide control (Sc-Tat), attenuated acetylation of p53 and reduced levels of Puma following GCI. CONCLUSIONS/SIGNIFICANCE: The studies demonstrate that p53 undergoes enhanced acetylation in the hippocampal CA1 region following global cerebral ischemia, and that the neuroprotective agent, 17ß-E(2), markedly attenuates the ischemia-induced p53 acetylation. Furthermore, following LTED, the suppressive effect of 17ß-E(2) on p53 acetylation is lost, and p53 acetylation increases in the hippocampus, which may explain previous reports of increased sensitivity of the hippocampus to ischemic stress following LTED.


Subject(s)
Brain Ischemia/metabolism , Estrogens/pharmacology , Hippocampus/metabolism , Tumor Suppressor Protein p53/metabolism , Acetylation , Animals , Female , Neuroprotective Agents , Rats , Rats, Sprague-Dawley , p300-CBP Transcription Factors
17.
Proc Natl Acad Sci U S A ; 108(35): E617-24, 2011 Aug 30.
Article in English | MEDLINE | ID: mdl-21808025

ABSTRACT

Recent work suggests that timing of 17ß-estradiol (E2) therapy may be critical for observing a beneficial neural effect. Along these lines, E2 neuroprotection, but not its uterotropic effect, was shown to be lost following long-term E2 deprivation (LTED), and this effect was associated with a significant decrease of estrogen receptor-α (ERα) in the hippocampus but not the uterus. The purpose of the current study was to determine the mechanism underlying the ERα decrease and to determine whether aging leads to a similar loss of hippocampal ERα and E2 sensitivity. The results of the study show that ERα in the rat hippocampal CA1 region but not the uterus undergoes enhanced interaction with the E3 ubiquitin ligase C terminus of heat shock cognate protein 70 (Hsc70)-interacting protein (CHIP) that leads to its ubiquitination/proteasomal degradation following LTED (10-wk ovariectomy). E2 treatment initiated before but not after LTED prevented the enhanced ERα-CHIP interaction and ERα ubiquitination/degradation and was fully neuroprotective against global cerebral ischemia. Administration of a proteasomal inhibitor or CHIP antisense oligonucleotides to knock down CHIP reversed the LTED-induced down-regulation of ERα. Further work showed that these observations extended to natural aging, because aged rats showed enhanced CHIP interaction; ubiquitination and degradation of both hippocampal ERα and ERß; and, importantly, a correlated loss of E2 neuroprotection against global cerebral ischemia. In contrast, E2 administration to middle-aged rats was still capable of exerting neuroprotection. As a whole, the study provides support for a "critical period" for E2 neuroprotection of the hippocampus and provides important insight into the mechanism underlying the critical period.


Subject(s)
Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Hippocampus/metabolism , Neuroprotective Agents/pharmacology , Ubiquitin-Protein Ligases/physiology , Aging/metabolism , Animals , Female , Proteasome Endopeptidase Complex/physiology , Proteasome Inhibitors , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Ubiquitination
18.
PLoS One ; 5(9): e12606, 2010 Sep 07.
Article in English | MEDLINE | ID: mdl-20830300

ABSTRACT

BACKGROUND: Recent work by our laboratory and others has implicated NADPH oxidase as having an important role in reactive oxygen species (ROS) generation and neuronal damage following cerebral ischemia, although the mechanisms controlling NADPH oxidase in the brain remain poorly understood. The purpose of the current study was to examine the regulatory and functional role of the Rho GTPase, Rac1 in NADPH oxidase activation, ROS generation and neuronal cell death/cognitive dysfunction following global cerebral ischemia in the male rat. METHODOLOGY/PRINCIPAL FINDINGS: Our studies revealed that NADPH oxidase activity and superoxide (O(2)(-)) production in the hippocampal CA1 region increased rapidly after cerebral ischemia to reach a peak at 3 h post-reperfusion, followed by a fall in levels by 24 h post-reperfusion. Administration of a Rac GTPase inhibitor (NSC23766) 15 min before cerebral ischemia significantly attenuated NADPH oxidase activation and O(2)(-) production at 3 h after stroke as compared to vehicle-treated controls. NSC23766 also attenuated "in situ" O(2)(-) production in the hippocampus after ischemia/reperfusion, as determined by fluorescent oxidized hydroethidine staining. Oxidative stress damage in the hippocampal CA1 after ischemia/reperfusion was also significantly attenuated by NSC23766 treatment, as evidenced by a marked attenuation of immunostaining for the oxidative stress damage markers, 4-HNE, 8-OHdG and H2AX at 24 h in the hippocampal CA1 region following cerebral ischemia. In addition, Morris Water maze testing revealed that Rac GTPase inhibition after ischemic injury significantly improved hippocampal-dependent memory and cognitive spatial abilities at 7-9 d post reperfusion as compared to vehicle-treated animals. CONCLUSIONS/SIGNIFICANCE: The results of the study suggest that Rac1 GTPase has a critical role in mediating ischemia/reperfusion injury-induced NADPH oxidase activation, ROS generation and oxidative stress in the hippocampal CA1 region of the rat, and thus contributes significantly to neuronal degeneration and cognitive dysfunction following cerebral ischemia.


Subject(s)
Brain Ischemia/enzymology , Brain Ischemia/psychology , Cognition , NADPH Oxidases/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Brain Ischemia/metabolism , Disease Models, Animal , Enzyme Activation , Hippocampus/enzymology , Hippocampus/metabolism , Humans , Male , NADPH Oxidases/genetics , Oxidative Stress , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , rac1 GTP-Binding Protein/genetics
19.
PLoS One ; 5(5): e9851, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20479872

ABSTRACT

BACKGROUND: 17beta-estradiol (E2) has been implicated to exert neuroprotective effects in the brain following cerebral ischemia. Classically, E2 is thought to exert its effects via genomic signaling mediated by interaction with nuclear estrogen receptors. However, the role and contribution of extranuclear estrogen receptors (ER) is unclear and was the subject of the current study. METHODOLOGY/PRINCIPAL FINDINGS: To accomplish this goal, we employed two E2 conjugates (E2 dendrimer, EDC, and E2-BSA) that can interact with extranuclear ER and exert rapid nongenomic signaling, but lack the ability to interact with nuclear ER due to their inability to enter the nucleus. EDC or E2-BSA (10 microM) was injected icv 60 min prior to global cerebral ischemia (GCI). FITC-tagged EDC or E2-BSA revealed high uptake in the hippocampal CA1 region after icv injection, with a membrane (extranuclear) localization pattern in cells. Both EDC and E2-BSA exerted robust neuroprotection in the CA1 against GCI, and the effect was blocked by the ER antagonist, ICI182,780. EDC and E2-BSA both rapidly enhanced activation of the prosurvival kinases, ERK and Akt, while attenuating activation of the proapoptotic kinase, JNK following GCI, effects that were blocked by ICI182,780. Administration of an MEK or PI3K inhibitor blocked the neuroprotective effects of EDC and E2-BSA. Further studies showed that EDC increased p-CREB and BDNF in the CA1 region in an ERK- and Akt-dependent manner, and that cognitive outcome after GCI was preserved by EDC in an ER-dependent manner. CONCLUSIONS/SIGNIFICANCE: In conclusion, the current study demonstrates that activation of extranuclear ER results in induction of ERK-Akt-CREB-BDNF signaling in the hippocampal CA1 region, which significantly reduces ischemic neuronal injury and preserves cognitive function following GCI. The study adds to a growing literature that suggests that extranuclear ER can have important actions in the brain.


Subject(s)
Cell Nucleus/metabolism , Estrogens/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Neuroprotective Agents/pharmacology , Receptors, Estrogen/metabolism , Animals , Brain Ischemia/enzymology , Brain Ischemia/pathology , Brain-Derived Neurotrophic Factor/metabolism , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/enzymology , CA1 Region, Hippocampal/pathology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Nucleus/drug effects , Cell Survival/drug effects , Cognition/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Dendrimers/pharmacology , Estradiol/metabolism , Estradiol/pharmacology , Estrogens, Conjugated (USP)/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Injections, Intraventricular , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , Neurons/drug effects , Neurons/enzymology , Neurons/pathology , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Serum Albumin, Bovine/metabolism , Serum Albumin, Bovine/pharmacology
20.
Hippocampus ; 20(1): 65-77, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19330848

ABSTRACT

alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) are responsible for excitotoxicity induced by ischemic injury in hippocampal CA1 neurons, whereas the molecular mechanisms responsible for their neurotrophic activities are much less studied. Here, we examined the neuroprotective effect of positive modeulation of AMPARs by coapplication of AMPA with PEPA, an allosteric potentiator of AMPARs. We showed that coapplication of AMPA with PEPA protected hippocampal CA1 neurons from brain ischemia-induced death. Coapplication of AMPA with PEPA could prevent downregulated expression of GluR2 subunit caused by ischemia and increase BDNF expression via Lyn-ERK1/2-CREB signaling. Furthermore, TrkB receptor-mediated PI3K/Akt signal pathway was activated after coapplication of AMPA with PEPA, which was related to MAPK pathway and protected CA1 neurons against ischemic insults through depression of JNK3 activity, release of cytochrome c to cytosol and depression of capase-3 activity. Our results revealed that positive modulation of AMPARs could exert neuroprotective effects and the possible signaling pathways underlied.


Subject(s)
Brain Ischemia/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Receptors, AMPA/metabolism , src-Family Kinases/metabolism , Animals , CA1 Region, Hippocampal/metabolism , Gene Expression Regulation , MAP Kinase Signaling System , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/metabolism , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Receptors, AMPA/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL