Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Int J Mol Sci ; 21(8)2020 Apr 21.
Article in English | MEDLINE | ID: mdl-32326309

ABSTRACT

The association between plasma cholesterol levels and the development of dementia continues to be an important topic of discussion in the scientific community, while the results in the literature vary significantly. We study the effect of reducing oxidized neuronal cholesterol on the lipid raft structure of plasma membrane. The levels of plasma membrane cholesterol were reduced by treating the intact cells with methyl-ß-cyclodextrin (MßCD). The relationship between the cell viability with varying levels of MßCD was then examined. The viability curves are well described by a modified form of the empirical Gompertz law of mortality. A detailed statistical analysis is performed on the fitting results, showing that increasing MßCD concentration has a minor, rather than significant, effect on the cellular viability. In particular, the dependence of viability on MßCD concentration was found to be characterized by a ~25% increase per 1 µM of MßCD concentration.


Subject(s)
Cell Death , Cholesterol/metabolism , Neurons/metabolism , Stress, Physiological , Cell Membrane/metabolism , Cell Survival , Cells, Cultured , Cholesterol/pharmacology , Dose-Response Relationship, Drug , Neurons/drug effects
2.
J Am Chem Soc ; 138(50): 16549-16552, 2016 12 21.
Article in English | MEDLINE | ID: mdl-27998093

ABSTRACT

This paper describes the design and synthesis of a conjugate (Q7R) comprising the synthetic host cucurbit[7]uril (Q7) linked to the fluorescent dye tetramethylrhodamine (TMR), and the characterization of its optical and guest-binding properties as well as its cellular uptake. Q7R was synthesized in two steps from monofunctionalized azidobutyl-Q7 and NHS-activated TMR. The fluorescence of Q7R is quenched upon guest binding, and this observable was used to determine equilibrium dissociation constant (Kd) values. Unexpectedly, the Kd values for guests binding to Q7R and to unmodified Q7 were essentially identical. Therefore, Q7R can directly report binding to Q7 without an energetic penalty due to the conjugated fluorophore. This result demonstrates a potentially general strategy for the design of single-component host-indicator conjugates that respond sensitively to analytes without perturbing the binding properties of the host. The unique properties of Q7R enabled measurement of Kd values across 3 orders of magnitude and at concentrations as low as 0.7 nM. This result is particularly relevant given the unmatched range of guests and binding affinities demonstrated for Q7. Confocal fluorescence microscopy of live and fixed HT22 neurons revealed the cellular uptake of Q7R and its punctate localization in the cytoplasm. Q7R did not alter cell growth at concentrations up to 2.2 µM over 4 days. These experiments demonstrate the feasibility of Q7R as a direct sensor for guest binding and as a cell-permeable compound for imaging applications.


Subject(s)
Bridged-Ring Compounds/chemistry , Imidazoles/chemistry , Molecular Imaging/methods , Rhodamines/chemistry , Cell Line, Tumor , Humans
3.
Mol Cell Proteomics ; 13(3): 760-79, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24390426

ABSTRACT

Recent clinical evidence suggests that the neuroprotective and beneficial effects of hormone therapy may be limited by factors related to age and reproductive status. The patient's age and length of time without circulating ovarian hormones are likely to be key factors in the specific neurological outcomes of hormone therapy. However, the mechanisms underlying age-related changes in hormone efficacy have not been determined. We hypothesized that there are intrinsic changes in estrogen receptor ß (ERß) function that determine its ability to mediate the actions of 17ß-estradiol (E2) in brain regions such as the ventral hippocampus. In this study, we identified and quantified a subset of ERß protein interactions in the ventral hippocampus that were significantly altered by E2 replacement in young and aged animals, using two-dimensional differential gel electrophoresis coupled with liquid chromatography-electrospray ionization-tandem mass spectrometry. This study demonstrates quantitative changes in ERß protein-protein interactions with E2 replacement that are dependent upon age in the ventral hippocampus and how these changes could alter processes such as transcriptional regulation. Thus, our data provide evidence that changes in ERß protein interactions are a potential mechanism for age-related changes in E2 responsiveness in the brain after menopause.


Subject(s)
Aging/metabolism , Estradiol/pharmacology , Estrogen Receptor beta/metabolism , Hippocampus/metabolism , Protein Interaction Mapping , Adenosine Triphosphatases/metabolism , Aging/drug effects , Animals , Annexin A5/metabolism , Cell Cycle Proteins/metabolism , Estrogen Receptor beta/genetics , Female , Gelsolin/metabolism , Gene Knockdown Techniques , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , HEK293 Cells , Hippocampus/drug effects , Humans , Image Processing, Computer-Assisted , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , RNA, Small Interfering/metabolism , Rats , Rats, Inbred F344 , Response Elements/genetics , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Transcription, Genetic/drug effects , Valosin Containing Protein
4.
J Pharmacol Exp Ther ; 349(3): 526-32, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24706985

ABSTRACT

Numerous studies have demonstrated that females have a higher risk of experiencing several pain disorders with either greater frequency or severity than males. Although the mechanisms that underlie this sex disparity remain unclear, several studies have shown an important role for sex steroids, such as estrogen, in the modulation of nociception. Receptors for estrogen are present in primary afferent neurons in the trigeminal and dorsal root ganglia, and brief exposure to estrogen increases responses to the inflammatory mediator bradykinin (BK). However, the mechanism for estrogen-mediated enhancement of BK signaling is not fully understood. The aim of the present study was to evaluate the relative contributions of estrogen receptor α (ERα), ERß, and G protein-coupled estrogen receptor 1 (GPER) to the enhanced signaling of the inflammatory mediator BK by 17ß-estradiol (17ß-E2) in primary sensory neurons from female rats in culture (ex vivo) and in behavioral assays of nociception in vivo. The effects of 17ß-E2 on BK responses were mimicked by ERα-selective agonists and blocked by ERα-selective antagonists and by small interfering RNA knockdown of ERα. The data indicate that ERα is required for 17ß-E2-mediated enhancement of BK signaling in peripheral sensory neurons in female rats.


Subject(s)
Bradykinin/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Sensory Receptor Cells/drug effects , Signal Transduction/drug effects , Trigeminal Ganglion/drug effects , Animals , Behavior, Animal/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/antagonists & inhibitors , Female , Hyperalgesia/metabolism , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/metabolism , Sex Factors , Trigeminal Ganglion/cytology , Trigeminal Ganglion/metabolism
5.
Nat Med ; 13(1): 89-94, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17195839

ABSTRACT

Metabolic hormones, such as leptin, alter the input organization of hypothalamic circuits, resulting in increased pro-opiomelanocortin (POMC) tone, followed by decreased food intake and adiposity. The gonadal steroid estradiol can also reduce appetite and adiposity, and it influences synaptic plasticity. Here we report that estradiol (E2) triggers a robust increase in the number of excitatory inputs to POMC neurons in the arcuate nucleus of wild-type rats and mice. This rearrangement of synapses in the arcuate nucleus is leptin independent because it also occurred in leptin-deficient (ob/ob) and leptin receptor-deficient (db/db) mice, and was paralleled by decreased food intake and body weight gain as well as increased energy expenditure. However, estrogen-induced decrease in body weight was dependent on Stat3 activation in the brain. These observations support the notion that synaptic plasticity of arcuate nucleus feeding circuits is an inherent element in body weight regulation and offer alternative approaches to reducing adiposity under conditions of failed leptin receptor signaling.


Subject(s)
Estradiol/pharmacology , Melanocortins/metabolism , Neurons/drug effects , Obesity/physiopathology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Animals , Anorexia/chemically induced , Anorexia/physiopathology , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/physiology , Arcuate Nucleus of Hypothalamus/ultrastructure , Body Weight/drug effects , Estradiol/administration & dosage , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/physiology , Excitatory Postsynaptic Potentials/drug effects , Female , Injections, Intraventricular , Leptin/genetics , Leptin/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Microscopy, Electron , Neurons/cytology , Neurons/metabolism , Obesity/genetics , Ovariectomy , Pro-Opiomelanocortin/metabolism , Rats , Rats, Sprague-Dawley
6.
J Neurosci ; 31(1): 157-63, 2011 Jan 05.
Article in English | MEDLINE | ID: mdl-21209200

ABSTRACT

Mutations in parkin, an E3 ubiquitin ligase, are the most common cause of autosomal-recessive Parkinson's disease (PD). Here, we show that the stress-signaling non-receptor tyrosine kinase c-Abl links parkin to sporadic forms of PD via tyrosine phosphorylation. Under oxidative and dopaminergic stress, c-Abl was activated in cultured neuronal cells and in striatum of adult C57BL/6 mice. Activated c-Abl was found in the striatum of PD patients. Concomitantly, parkin was tyrosine-phosphorylated, causing loss of its ubiquitin ligase and cytoprotective activities, and the accumulation of parkin substrates, AIMP2 (aminoacyl tRNA synthetase complex-interacting multifunctional protein 2) (p38/JTV-1) and FBP-1.STI-571, a selective c-Abl inhibitor, prevented tyrosine phosphorylation of parkin and restored its E3 ligase activity and cytoprotective function both in vitro and in vivo. Our results suggest that tyrosine phosphorylation of parkin by c-Abl is a major post-translational modification that leads to loss of parkin function and disease progression in sporadic PD. Moreover, inhibition of c-Abl offers new therapeutic opportunities for blocking PD progression.


Subject(s)
Gene Expression Regulation/physiology , MPTP Poisoning/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Tyrosine/metabolism , Ubiquitin-Protein Ligases/metabolism , Acetylcysteine/pharmacology , Animals , Benzamides , Brain/drug effects , Brain/metabolism , Brain/pathology , Case-Control Studies , Cell Line , Disease Models, Animal , Dopamine/pharmacology , Drug Administration Schedule , Free Radical Scavengers/pharmacology , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/genetics , Humans , Imatinib Mesylate , Immunoprecipitation/methods , MPTP Poisoning/chemically induced , MPTP Poisoning/drug therapy , MPTP Poisoning/pathology , Male , Metalloporphyrins/pharmacology , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Oxidative Stress/physiology , Peptide Elongation Factors/genetics , Peptide Elongation Factors/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , Piperazines/toxicity , Polyethylene Glycols/pharmacology , Protein Kinase Inhibitors/toxicity , Proto-Oncogene Proteins c-abl/genetics , Pyrimidines/toxicity , RNA, Small Interfering/pharmacology , Statistics, Nonparametric , Transfection/methods , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination/drug effects
7.
Mol Cell Neurosci ; 47(3): 181-90, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21545837

ABSTRACT

The proteasome is an enzyme complex responsible for targeted intracellular proteolysis. Alterations in proteasome-mediated protein clearance have been implicated in the pathogenesis of aging, Alzheimer's disease (AD) and Parkinson's disease (PD). In such diseases, proteasome inhibition may contribute to formation of abnormal protein aggregates, which in turn activate intracellular unfolded protein responses that cause oxidative stress and apoptosis. In this study, we investigated the protective effect of Insulin-like Growth Factor-I (IGF-1) for neural SH-SY5Y cells treated with the proteasomal inhibitor, Epoxomicin. In SH-SY5Y cells, Epoxomicin treatment results in accumulation of intracellular ubiquitinated proteins and cytochrome c release from damaged mitochondria, leading to cell death, in Epoxomicin time- and dose-dependent manner. In cells treated with small amounts of IGF-1, the same dosages of Epoxomicin reduced both mitochondrial damage (cytochrome c release) and reduced caspase-3 activation and PARP cleavage, both of which are markers of apoptosis. Notably, however, IGF-1-treated SH-SY5Y cells still contained ubiquitinated protein aggregates. This result indicates that IGF-1 blocks the downstream apoptotic consequences of Epoxomicin treatment leading to decreased proteasome function. Clues as to the mechanism for this protective effect come from (a) increased AKT phosphorylation observed in IGF-1-protected cells, vs. cells exposed to Epoxomicin without IGF-1, and (b) reduction of IGF-1 protection by pretreatment of the cells with LY294002 (an inhibitor of PI3-kinase). Together these findings suggest that activation of PI3/AKT pathways by IGF-1 is involved in IGF-1 neuroprotection against apoptosis following proteasome inhibition.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Mitochondria/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Proteasome Inhibitors , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cytoprotection/physiology , Dose-Response Relationship, Drug , Humans , Insulin-Like Growth Factor I/metabolism , Mitochondria/metabolism , Neurons/metabolism , Neuroprotective Agents/metabolism , Oligopeptides/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
8.
Horm Behav ; 60(5): 617-24, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21907204

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by dopamine neuron loss in the nigrostriatal pathway that shows greater incidence in men than women. The mechanisms underlying this gender bias remain elusive, although one possibility is that androgens may increase dopamine neuronal vulnerability to oxidative stress. Motor impairment can be modeled in rats receiving a unilateral injection of 6-hydroxydopamine (6-OHDA), a neurotoxin producing nigrostriatal degeneration. To investigate the role of androgens in PD, we compared young (2 months) and aged (24 months) male rats receiving gonadectomy (GDX) and their corresponding intact controls. One month after GDX, rats were unilaterally injected with 6-OHDA, and their motor impairment and asymmetry were assessed 2 weeks later using the cylinder test and the amphetamine-induced rotation test. Plasma samples were also collected to assess the concentration of testosterone and advanced oxidation protein products, a product of oxidative stress. GDX decreased lesion-induced asymmetry along with oxidative stress and increased amphetamine-induced rotations. These results show that GDX improves motor behaviors by decreasing motor asymmetry in 6-OHDA-treated rats, an effect that may be ascribed to increased release of striatal dopamine and decreased oxidative stress. Collectively, the data support the hypothesis that androgens may underlie the gender bias observed in PD.


Subject(s)
Adrenergic Agents/adverse effects , Androgens/pharmacology , Dopaminergic Neurons/drug effects , Motor Activity/drug effects , Neostriatum/drug effects , Oxidopamine/adverse effects , Substantia Nigra/drug effects , Amphetamine/pharmacology , Animals , Male , Orchiectomy , Oxidative Stress/drug effects , Rats , Rotation , Testosterone/blood
9.
Mol Ther ; 18(8): 1536-44, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20531393

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) has emerged as the most potent neuroprotective agent tested in experimental models for the treatment of Parkinson's disease (PD). However, its use is hindered by difficulties in delivery to the brain due to the presence of the blood-brain barrier (BBB). In order to circumvent this problem, we took advantage of the fact that bone marrow stem cell-derived macrophages are able to pass the BBB and home to sites of neuronal degeneration. Here, we report the development of a method for brain delivery of GDNF by genetically modified macrophages. Bone marrow stem cells were transduced ex vivo with lentivirus expressing a GDNF gene driven by a synthetic macrophage-specific promoter and then transplanted into recipient mice. Eight weeks after transplantation, the mice were injected with the neurotoxin, MPTP, for 7 days to induce dopaminergic neurodegeneration. Macrophage-mediated GDNF treatment dramatically ameliorated MPTP-induced degeneration of tyrosine hydroxylase (TH)-positive neurons of the substantia nigra and TH(+) terminals in the striatum, stimulated axon regeneration, and reversed hypoactivity in the open field test. These results indicate that macrophage-mediated GDNF delivery is a promising strategy for developing a neuroprotective therapy for PD.


Subject(s)
Dopamine/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Macrophages/metabolism , Nerve Degeneration/therapy , Parkinson Disease/therapy , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Body Weight/drug effects , Cells, Cultured , Chromatography, High Pressure Liquid , Eating/drug effects , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Glial Cell Line-Derived Neurotrophic Factor/genetics , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/chemically induced , Neurotoxins/pharmacology , Parkinson Disease/metabolism , Substantia Nigra/drug effects , Substantia Nigra/metabolism
10.
J Pharmacol Exp Ther ; 335(1): 190-6, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20647494

ABSTRACT

Many studies have demonstrated that premenopausal women are at increased risk for various pain disorders. Pain-sensing neurons, termed "nociceptors," in the trigeminal ganglia (TG) and dorsal root ganglia (DRG) express receptors for inflammatory mediators and noxious physical stimuli and transmit signals for central processing of pain sensation. Estrogen receptors (ERs) are also expressed on nociceptors in the TG and DRG, and there is ample literature to suggest that activation of ERs can influence pain mechanisms. However, the mechanism for ER modulation of nociceptor activity is incompletely understood. The aim of this study was to characterize the effect of 17ß-estradiol (17ß-E(2)) on signaling of the inflammatory mediator bradykinin (BK) in primary cultures of rat sensory neurons and a behavioral model of thermal allodynia in rats. Here, we show that exposure to 17ß-E(2) rapidly (within 15 min) enhanced responses to BK in vitro and in vivo. The 17ß-E(2)-mediated enhancement of BK signaling was not blocked by the transcription inhibitor anisomycin and was mediated by a membrane-associated ER. The effect of 17ß-E(2) to enhance BK responses required activation of ß1-containing, RGD-binding integrins. These data show that 17ß-E(2) rapidly enhances inflammatory mediator responses both in vitro and in vivo and suggest that 17ß-E(2) acting at primary sensory pain neurons may participate in regulating the sensitivity of women to painful stimuli.


Subject(s)
Bradykinin/physiology , Estradiol/pharmacology , Sensory Receptor Cells/physiology , Signal Transduction/drug effects , Animals , Anisomycin/pharmacology , Behavior, Animal/drug effects , Cells, Cultured , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Inositol Phosphates/metabolism , Integrins/antagonists & inhibitors , Integrins/metabolism , Male , Nerve Endings/drug effects , Nucleic Acid Synthesis Inhibitors/pharmacology , Pain/psychology , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/drug effects , Trigeminal Ganglion/cytology , Trigeminal Ganglion/drug effects , Type C Phospholipases/metabolism
11.
Eur J Neurosci ; 29(11): 2177-86, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19490092

ABSTRACT

Parkinson's disease (PD) is characterized by the progressive loss of nigrostriatal dopamine neurons leading to motor disturbances and cognitive impairment. Current pharmacotherapies relieve PD symptoms temporarily but fail to prevent or slow down the disease progression. In this study, we investigated the molecular mechanisms by which the non-selective cannabinoid receptor agonist WIN55,212-2 (WIN) protects mouse nigrostriatal neurons from 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurotoxicity and neuroinflammation. Stereological analyses showed that chronic treatment with WIN (4 mg/kg, intraperitoneal), initiated 24 h after MPTP administration, protected against MPTP-induced loss of tyrosine hydroxylase-positive neurons in the substantia nigra pars compacta independently of CB1 cannabinoid receptor activation. The neuroprotective effect of WIN was accompanied by increased dopamine and 3,4-dihydroxyphenylacetic acid levels in the substantia nigra pars compacta and dorsal striatum of MPTP-treated mice. At 3 days post-MPTP, we found significant microglial activation and up-regulation of CB2 cannabinoid receptors in the ventral midbrain. Treatment with WIN or the CB2 receptor agonist JWH015 (4 mg/kg, intraperitoneal) reduced MPTP-induced microglial activation, whereas genetic ablation of CB2 receptors exacerbated MPTP systemic toxicity. Furthermore, chronic WIN reversed MPTP-associated motor deficits, as revealed by the analysis of forepaw step width and percentage of faults using the inverted grid test. In conclusion, our data indicate that agonism at CB2 cannabinoid receptors protects against MPTP-induced nigrostriatal degeneration by inhibiting microglial activation/infiltration and suggest that CB2 receptors represent a new therapeutic target to slow the degenerative process occurring in PD.


Subject(s)
Benzoxazines/pharmacology , Cannabinoid Receptor Agonists , Disease Models, Animal , MPTP Poisoning/prevention & control , Morpholines/pharmacology , Naphthalenes/pharmacology , Neuroprotective Agents/pharmacology , Parkinson Disease, Secondary/prevention & control , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Cannabinoids/pharmacology , Cannabinoids/therapeutic use , Cell Count , Corpus Striatum/drug effects , Corpus Striatum/pathology , MPTP Poisoning/chemically induced , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/therapeutic use , Parkinson Disease, Secondary/chemically induced , Receptors, Cannabinoid/physiology , Substantia Nigra/drug effects , Substantia Nigra/pathology
12.
Trends Endocrinol Metab ; 18(10): 386-92, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17997103

ABSTRACT

Luteinizing hormone-releasing hormone-I (LHRH-I) was isolated from the mammalian hypothalamus and shown to be the primary regulator of reproduction through its initiation of pituitary gonadotropin release. Subsequently, it has also been shown to have non-pituitary actions. Although the regulation of LHRH-I synthesis and release has been extensively studied, there is additional evidence to suggest that processing of the peptide represents another layer of regulation. The focus of this review will be on evidence for the action of LHRH-(1-5), the pentapeptide metabolite of LHRH-I, in regulating LHRH-I synthesis, secretion and reproductive behavior. The involvement of LHRH-(1-5) in the control of aspects of reproduction might represent yet another level of regulatory complexity through neuropeptide processing.


Subject(s)
Gonadotropin-Releasing Hormone/physiology , Peptide Fragments/physiology , Reproduction/physiology , Animals , Brain/physiology , Feedback, Physiological , Female , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/biosynthesis , Gonadotropin-Releasing Hormone/metabolism , Homeostasis , Humans , Male , Metalloendopeptidases/analysis , Metalloendopeptidases/metabolism , Peptide Fragments/biosynthesis , Pituitary Gland/physiology , Pyrrolidonecarboxylic Acid/analogs & derivatives , Pyrrolidonecarboxylic Acid/metabolism
13.
Endocrinology ; 147(5): 2544-9, 2006 May.
Article in English | MEDLINE | ID: mdl-16497796

ABSTRACT

In the female rat, ovulation is preceded by a marked increase in the release of the decapeptide, LHRH, culminating in a preovulatory LH surge, which coincides with a period of sexual receptivity. The decapeptide, LHRH, is processed by a zinc metalloendopeptidase EC 3.4.24.15 (EP24.15) that cleaves the hormone at the Tyr(5)-Gly(6) bond. We have previously reported that the autoregulation of LHRH gene expression can also be mediated by its metabolite, LHRH-(1-5). Given the central function of LHRH in reproduction and reproductive behavior, we examined the role of the metabolite, LHRH-(1-5), in mediation of LHRH-facilitated reproductive behavior. Intracerebroventricular administration of LHRH-(1-5) facilitated sexual behavior responses, similar to those facilitated by the decapeptide LHRH, in ovariectomized estradiol-primed female rats. Furthermore, immunoneutralization of EP24.15 resulted in the inhibition of the LHRH-facilitated lordosis but had no inhibitory effects on LHRH-(1-5)-facilitated lordosis. The LHRH antagonist, Antide, was capable of inhibiting LHRH-facilitated lordosis, without affecting LHRH-(1-5)-facilitated lordosis. Collectively, these results suggest a role for LHRH metabolites in the facilitation of female receptive behavior in rats.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Posture , Sexual Behavior, Animal , Analysis of Variance , Animals , Behavior, Animal , Catheterization , Estradiol/metabolism , Female , Gene Expression Regulation , Luteinizing Hormone/metabolism , Nervous System Physiological Phenomena , Neurons/metabolism , Oligopeptides/pharmacology , Ovariectomy , Ovulation , Peptides/chemistry , Rabbits , Radiosurgery , Rats , Rats, Sprague-Dawley
14.
Endocrinology ; 147(4): 1924-31, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16439454

ABSTRACT

GnRH is the most upstream regulator of reproduction in vertebrates, and its synthesis and release are regulated by gonadal steroid hormones. The proposed sites of hormone action were historically thought to be upstream from GnRH neurons; however, the discovery of ERbeta in a subset of GnRH neurons suggests that this hypothesis should be reevaluated. To determine a functional role for ERbeta in GnRH neurons, we examined ERbeta's regulation of GnRH promoter activity. The GnRH-producing cell line, GT1-7, was cotransfected with expression vectors containing one of three ERbeta splice variants and a luciferase-reporter construct containing the full-length mouse GnRH promoter sequence or one of two deletions upstream of the transcription start site (-225/-201; -184/-150). Transfected cells were treated with 100 nm 17beta-estradiol (E(2)), diarylpropionitrile, raloxifene, or vehicle. There was a robust increase in GnRH-luciferase activity by all ERbeta splice variants in the absence of hormone. Furthermore, E(2) treatment abolished this response for ER-beta1 and ER-beta2, but not ER-beta1delta3. The -225/-201 and -184/-150 regions were critical for ERbeta-induced promoter activity because deletion of these regions eliminated the ligand-independent effects of ERbeta. ER-beta1 binds directly to these promoter regions and because there are no classical estrogen response elements in the mouse GnRH promoter, these data raise the possibility that this region contains a novel estrogen response element specific for ERbeta. Overall, our data suggest that ERbeta functions as a basic transcription factor in GnRH neurons and demonstrate a potential molecular mechanism for the negative feedback effects of E(2) on GnRH.


Subject(s)
Estrogen Receptor beta/physiology , Gonadotropin-Releasing Hormone/genetics , Promoter Regions, Genetic , Alternative Splicing , Animals , Base Sequence , Binding Sites , Cell Line , Cyclic AMP/biosynthesis , Estrogen Receptor beta/genetics , Feedback, Physiological , Ligands , Mice , Molecular Sequence Data
15.
Neurosci Lett ; 610: 79-85, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26520464

ABSTRACT

Astrocytes regulate neuronal homeostasis and have been implicated in affecting the viability and functioning of surrounding neurons under stressed and injured conditions. Previous data from our lab suggests indirect actions of estrogen through ERα in neighboring astroglia to protect dopamine neurons against 1-methyl-4-phenylpyridinium (MPP(+)) toxicity in mouse mesencephalic cultures. We further evaluate estrogen signaling in astrocytes and the mechanism of estrogen's indirect neuroprotective effects on dopamine neurons. Primary mesencephalic cultures pre-treated with 17ß-estradiol and the membrane impermeable estrogen, E2-BSA, were both neuroprotective against MPP(+) -induced dopamine neuron toxicity, suggesting membrane-initiated neuroprotection. ERα was found in the plasma membrane of astrocyte cultures and colocalized with the lipid raft marker, flotillin-1. A 17ß-estradiol time course revealed a significant increase in Akt, which was inhibited by the PI3 kinase inhibitor, LY294004. Estrogen conditioned media collected from pure astrocyte cultures rescued glial deficient mesencephalic cultures from MPP(+). This indirect estrogen-mediated neuroprotective effect in mesencephalic cultures was significantly reduced when PI3 kinase signaling in astrocytes was blocked prior to collecting estrogen-conditioned media using the irreversible PI3 kinase inhibitor, Wortmannin. Estrogen signaling via astrocytes is rapidly initiated at the membrane level and requires PI3 kinase signaling in order to protect primary mesencephalic dopamine neurons from MPP(+) neurotoxicity.


Subject(s)
Astrocytes/drug effects , Dopaminergic Neurons/drug effects , Estradiol/pharmacology , Estrogens/pharmacology , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , 1-Methyl-4-phenylpyridinium/toxicity , Androstadienes/pharmacology , Animals , Astrocytes/metabolism , Cell Membrane/metabolism , Culture Media, Conditioned , Dopaminergic Neurons/cytology , Estrogen Receptor alpha/metabolism , Mesencephalon/cytology , Mice, Inbred C57BL , Phosphoinositide-3 Kinase Inhibitors , Primary Cell Culture , Serum Albumin, Bovine/pharmacology , Signal Transduction , Wortmannin
16.
Endocrinology ; 146(1): 280-6, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15486221

ABSTRACT

Given the central role of the decapeptide LHRH in reproduction and reproductive behavior, it is important to focus on delineating the possible effects of this gene and its products in the regulation of hormone-dependent reproductive processes. In the female, ovulation is preceded by a marked increase in LHRH release; the increase in LHRH release culminates in a preovulatory LH surge, which coincides with a period of sexual receptivity. In contrast to the belief that the proteolytic metabolism of LHRH serves only as a degradative process that removes excess LHRH and attenuates signal transduction through the LHRH receptor, we hypothesized that a metabolite of the decapeptide, LHRH-(1-5), can directly regulate LHRH neuronal function. This study demonstrates the ability of LHRH-(1-5) peptide to regulate LHRH gene expression in the LHRH neuronal cell line, the GT(1-7) cell. The results show that LHRH-(1-5) stimulated LHRH gene expression at the posttranscriptional level. In contrast to the LHRH suppression of its own gene expression, the coadministration of LHRH with the metalloendopeptidase, EC 3.4.24.15, an endopeptidase known to cleave LHRH to form LHRH(1-5), shows a reversal of effect, a stimulation of LHRH gene expression. Finally, the effect of LHRH-(1-5) on LHRH gene expression appears to be mediated by the calcium/calmodulin-dependent protein kinase. The present study supports the hypothesis that the physiological metabolite of LHRH, LHRH-(1-5), is functionally capable of regulating the reproductive neuroendocrine system.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/pharmacology , Neurons/metabolism , Peptide Fragments/pharmacology , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line, Transformed , Gene Expression/drug effects , Gonadotropin-Releasing Hormone/genetics , Mice , Neurons/drug effects
17.
Diabetes ; 52(11): 2675-83, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14578285

ABSTRACT

Hypothalamic proopiomelanocortin (POMC) gene expression is reduced in many forms of obesity and diabetes, particularly in those attributable to deficiencies in leptin or its receptor. To assess the functional significance of POMC in mediating metabolic phenotypes associated with leptin deficiency, leptin-deficient mice bearing a transgene expressing the POMC gene under control of the neuron-specific enolase promoter were produced. The POMC transgene attenuated fasting-induced hyperphagia in wild-type mice. Furthermore, the POMC transgene partially reversed obesity, hyperphagia, and hypothermia and effectively normalized hyperglycemia, glucosuria, glucose intolerance, and insulin resistance in leptin-deficient mice. Effects of the POMC transgene on glucose homeostasis were independent of the partial correction of hyperphagia and obesity. Furthermore, the POMC transgene normalized the profile of hepatic and adipose gene expression associated with gluconeogenesis, glucose output, and insulin sensitivity. These results indicate that central POMC is a key modulator of glucose homeostasis and that agonists of POMC products may provide effective therapy in treating impairments in glucose homeostasis when hypothalamic POMC expression is reduced, as occurs with leptin deficiency, hypothalamic damage, and aging.


Subject(s)
Fasting/physiology , Hyperphagia/prevention & control , Leptin/deficiency , Neurons/physiology , Obesity/genetics , Pro-Opiomelanocortin/genetics , Adipose Tissue/anatomy & histology , Animals , Base Sequence , Body Weight , Cloning, Molecular , DNA Primers , Glucose Tolerance Test , Insulin/blood , Leptin/genetics , Leptin/physiology , Mice , Mice, Transgenic , Phosphopyruvate Hydratase/genetics , Pro-Opiomelanocortin/physiology
18.
FEBS J ; 272(12): 2978-92, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15955058

ABSTRACT

The metalloendopeptidase 24.15 (EP24.15) is ubiquitously present in the extracellular environment as a secreted protein. Outside the cell, this enzyme degrades several neuropeptides containing from 5 to 17 amino acids (e.g. gonadotropin releasing hormone, bradykinin, opioids and neurotensin). The constitutive secretion of EP24.15 from glioma C6 cells was demonstrated to be stimulated linearly by reduced concentrations of extracellular calcium. In the present report we demonstrate that extracellular calcium concentration has no effect on the total amount of the extracellular (cell associated + medium) enzyme. Indeed, immuno-cytochemical analyses by confocal and electron microscopy suggested that the absence of calcium favors the enzyme shedding from the plasma membrane into the medium. Two putative calcium-binding sites on EP24.15 (D93 and D159) were altered by site-directed mutagenesis to investigate their possible contribution to binding of the enzyme at the cell surface. These mutated recombinant proteins behave similarly to the wild-type enzyme regarding enzymatic activity, secondary structure, calcium sensitivity and immunoreactivity. However, immunocytochemical analyses by confocal microscopy consistently show a reduced ability of the D93A mutant to associate with the plasma membrane of glioma C6 cells when compared with the wild-type enzyme. These data and the model of the enzyme's structure as determined by X-ray diffraction suggest that D93 is located at the enzyme surface and is consistent with membrane association of EP24.15. Moreover, calcium was also observed to induce a major change in the EP24.15 cleavage site on distinctive fluorogenic substrates. These data suggest that calcium may be an important modulator of ep24.15 cell function.


Subject(s)
Calcium/metabolism , Cell Membrane/metabolism , Metalloendopeptidases/chemistry , Metalloendopeptidases/metabolism , Animals , Binding Sites , Cell Line, Tumor , Central Nervous System Neoplasms/enzymology , Circular Dichroism , Glioma/enzymology , Metalloendopeptidases/genetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Structure, Secondary , Rats , Substrate Specificity
19.
Brain Res ; 985(1): 1-12, 2003 Sep 19.
Article in English | MEDLINE | ID: mdl-12957363

ABSTRACT

Hypothalamic POMC neurons mediate catabolic responses such as decreased food intake and increased energy expenditure by, in part, monitoring levels of metabolic factors such as glucose, insulin and leptin. Recently, fatty acid synthase inhibitors were reported to reduce body weight, inhibit food intake, and increase metabolic rate, possibly by acting on hypothalamic neurons through a mechanism involving malonyl-CoA accumulation. Given the observation that leptin mediates similar catabolic effects by, in part, activating hypothalamic POMC neurons, it is possible that other catabolic signals such as feeding and fatty acid synthase inhibition may also activate POMC neurons. To test this hypothesis, hypothalamic sections from mice that were fed or injected with the fatty acid synthase inhibitor cerulenin were examined for Fos (a marker for neuronal activation) and POMC product immunoreactivity and compared with similarly processed sections from leptin-injected mice. Feeding increased Fos immunoreactivity in the lateral peri-arcuate area of the hypothalamus of both wild-type and leptin-deficient ob/ob mice (P<0.05), indicating that nutritional activation of the hypothalamus can be leptin-independent. Furthermore, feeding significantly induced Fos immunoreactivity in neurons expressing POMC (P<0.003), indicating that feeding, like leptin, activates POMC neurons. Injection with cerulenin, like feeding and leptin, also increased Fos immunoreactivity in the lateral peri-arcuate area (P<0.03) and, more specifically, in neurons expressing POMC. In contrast, injection with cerulenin had no grossly observable effects on cortical Fos immunoreactivity and appeared to suppress fasting-induced Fos immunoreactivity by about 35% (although the decrease did not reach statistical significance) in the medial arcuate nucleus, an area associated with anabolic responses such as increased food intake. Injection with cerulenin also decreased Fos immunoreactivity in the granular layer of the dentate gyrus of the hippocampus by about 30% (P<0.05), further suggesting that cerulenin does not non-specifically activate wide varieties of neurons. These results suggest that activation of hypothalamic POMC neurons may help to mediate some of the catabolic effects associated with feeding, cerulenin and leptin.


Subject(s)
Antifungal Agents/pharmacology , Cerulenin/pharmacology , Eating/drug effects , Leptin/pharmacology , Neurons/drug effects , Animals , Eating/physiology , Hypothalamus/drug effects , Hypothalamus/physiology , Immunohistochemistry , Leptin/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains/genetics , Neurons/physiology , Pro-Opiomelanocortin/metabolism , Proto-Oncogene Proteins c-fos/metabolism
20.
Neurochem Int ; 62(1): 31-42, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23142153

ABSTRACT

Inhibition of proteasome activity and the resulting protein accumulation are now known to be important events in the development of many neurological disorders, including Alzheimer's and Parkinson's diseases. Abnormal or over expressed proteins cause endoplasmic reticulum and oxidative stress leading to cell death, thus, normal proteasome function is critical for their removal. We have shown previously, with cultured SH-SY5Y neuroblastoma cells, that proteasome inhibition by the drug epoxomicin results in accumulation of ubiquitinated proteins. This causes obligatory loading of the mitochondria with calcium (Ca(2+)), resulting in mitochondrial damage and cytochrome c release, followed by programmed cell death (PCD). In the present study, we demonstrate that all-trans-retinoic acid (RA) pretreatment of SH-SY5Y cells protects them from PCD death after subsequent epoxomicin treatment which causes proteasome inhibition. Even though ubiquitinated protein aggregates are present, there is no evidence to suggest that autophagy is involved. We conclude that protection by RA is likely by mechanisms that interfere with cell stress-PCD pathway that otherwise would result from protein accumulation after proteasome inhibition. In addition, although RA activates both the AKT and ERK phosphorylation signaling pathways, only pretreatment with LY294002, an inhibitor of PI3-kinase in the AKT pathway, removed the protective effect of RA from the cells. This finding implies that RA activation of the AKT signaling cascade takes precedence over its activation of ERK1/2 phosphorylation, and that this selective effect of RA is key to its protection of epoxomicin-treated cells. Taken together, these findings suggest that RA treatment of cultured neuroblastoma cells sets up conditions under which proteasome inhibition, and the resultant accumulation of ubiquitinated proteins, loses its ability to kill the cells and may likely play a therapeutic role in neurodegenerative diseases.


Subject(s)
Cell Death/drug effects , Oncogene Protein v-akt/physiology , Proteasome Endopeptidase Complex/drug effects , Signal Transduction/drug effects , Tretinoin/pharmacology , Anti-Bacterial Agents/pharmacology , Blotting, Western , Caspase 3/metabolism , Caspase Inhibitors/pharmacology , Cell Death/genetics , Cell Line , Chromones/pharmacology , Data Interpretation, Statistical , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique , Humans , Membrane Potential, Mitochondrial/drug effects , Microscopy, Confocal , Morpholines/pharmacology , Oligopeptides/pharmacology , Oncogene Protein v-akt/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proteasome Endopeptidase Complex/genetics
SELECTION OF CITATIONS
SEARCH DETAIL