Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Endocrinology ; 163(1)2022 01 01.
Article in English | MEDLINE | ID: mdl-34919671

ABSTRACT

MicroRNAs (miRNAs) expressed in the hypothalamus are capable of regulating energy balance and peripheral metabolism by inhibiting translation of target messenger RNAs (mRNAs). Hypothalamic insulin resistance is known to precede that in the periphery, thus a critical unanswered question is whether central insulin resistance creates a specific hypothalamic miRNA signature that can be identified and targeted. Here we show that miR-1983, a unique miRNA, is upregulated in vitro in 2 insulin-resistant immortalized hypothalamic neuronal neuropeptide Y-expressing models, and in vivo in hyperinsulinemic mice, with a concomitant decrease of insulin receptor ß subunit protein, a target of miR-1983. Importantly, we demonstrate that miR-1983 is detectable in human blood serum and that its levels significantly correlate with blood insulin and the homeostatic model assessment of insulin resistance. Levels of miR-1983 are normalized with metformin exposure in mouse hypothalamic neuronal cell culture. Our findings provide evidence for miR-1983 as a unique biomarker of cellular insulin resistance, and a potential therapeutic target for prevention of human metabolic disease.


Subject(s)
Hypothalamus/metabolism , Insulin/pharmacology , Metformin/pharmacology , MicroRNAs/genetics , Receptor, Insulin/genetics , Adult , Animals , Cell Line , Cells, Cultured , Female , Gene Expression Profiling/methods , Gene Expression Regulation/drug effects , Humans , Hypoglycemic Agents/pharmacology , Hypothalamus/cytology , Insulin/blood , Insulin/metabolism , Insulin Resistance/genetics , Male , Mice , MicroRNAs/blood , Middle Aged , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Obesity/blood , Obesity/genetics , Obesity/metabolism , Receptor, Insulin/metabolism
2.
Mol Metab ; 31: 124-137, 2020 01.
Article in English | MEDLINE | ID: mdl-31918914

ABSTRACT

OBJECTIVES: The incretin hormone glucagon-like peptide-1 (GLP-1) is secreted from intestinal L-cells upon nutrient intake. While recent evidence has shown that GLP-1 is released in a circadian manner in rats, whether this occurs in mice and if this pattern is regulated by the circadian clock remain to be elucidated. Furthermore, although circadian GLP-1 secretion parallels expression of the core clock gene Bmal1, the link between the two remains largely unknown. Secretagogin (Scgn) is an exocytotic SNARE regulatory protein that demonstrates circadian expression and is essential for insulin secretion from ß-cells. The objective of the current study was to establish the necessity of the core clock gene Bmal1 and the SNARE protein SCGN as essential regulators of circadian GLP-1 secretion. METHODS: Oral glucose tolerance tests were conducted at different times of the day on 4-hour fasted C57BL/6J, Bmal1 wild-type, and Bmal1 knockout mice. Mass spectrometry, RNA-seq, qRT-PCR and/or microarray analyses, and immunostaining were conducted on murine (m) and human (h) primary L-cells and mGLUTag and hNCI-H716 L-cell lines. At peak and trough GLP-1 secretory time points, the mGLUTag cells were co-stained for SCGN and a membrane-marker, ChIP was used to analyze BMAL1 binding sites in the Scgn promoter, protein interaction with SCGN was tested by co-immunoprecipitation, and siRNA was used to knockdown Scgn for GLP-1 secretion assay. RESULTS: C57BL/6J mice displayed a circadian rhythm in GLP-1 secretion that peaked at the onset of their feeding period. Rhythmic GLP-1 release was impaired in Bmal1 knockout (KO) mice as compared to wild-type controls at the peak (p < 0.05) but not at the trough secretory time point. Microarray identified SNARE and transport vesicle pathways as highly upregulated in mGLUTag L-cells at the peak time point of GLP-1 secretion (p < 0.001). Mass spectrometry revealed that SCGN was also increased at this time (p < 0.001), while RNA-seq, qRT-PCR, and immunostaining demonstrated Scgn expression in all human and murine primary L-cells and cell lines. The mGLUTag and hNCI-H716 L-cells exhibited circadian rhythms in Scgn expression (p < 0.001). The ChIP analysis demonstrated increased binding of BMAL1 only at the peak of Scgn expression (p < 0.01). Immunocytochemistry showed the translocation of SCGN to the cell membrane after stimulation at the peak time point only (p < 0.05), while CoIP showed that SCGN was pulled down with SNAP25 and ß-actin, but only the latter interaction was time-dependent (p < 0.05). Finally, Scgn siRNA-treated cells demonstrated significantly blunted GLP-1 secretion (p < 0.01) in response to stimulation at the peak time point only. CONCLUSIONS: These data demonstrate, for the first time, that mice display a circadian pattern in GLP-1 secretion, which is impaired in Bmal1 knockout mice, and that Bmal1 regulation of Scgn expression plays an essential role in the circadian release of the incretin hormone GLP-1.


Subject(s)
ARNTL Transcription Factors/metabolism , Circadian Clocks/genetics , Glucagon-Like Peptide 1/metabolism , Secretagogins/metabolism , ARNTL Transcription Factors/deficiency , ARNTL Transcription Factors/genetics , Animals , Female , Glucose Tolerance Test , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
3.
Endocrinology ; 160(1): 181-192, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30500912

ABSTRACT

Bisphenol A (BPA), a ubiquitous environmental endocrine disruptor, is considered an obesogen. However, its role in the hypothalamic control of energy balance remains largely unexplored. Because disruption of the circadian clock is tightly associated with metabolic consequences, we explored how BPA affects the components of the molecular circadian clock in the feeding-related neurons of the hypothalamus. In immortalized POMC and NPY/AgRP-expressing hypothalamic cell lines and primary culture, we describe how BPA significantly alters mRNA expression of circadian clock genes Bmal1,Per2, and Rev-Erbα. Furthermore, we use newly generated Bmal1-knockout (KO) hypothalamic cell lines to link the BPA-induced neuropeptide dysregulation to the molecular clock. Specifically, BPA increased Npy, Agrp, and Pomc mRNA expression in wild type hypothalamic cells, whereas the increase in Npy, but not Agrp or Pomc, was abolished in cell lines lacking BMAL1. In line with this increase, BPA led to increased BMAL1 binding to the Npy promotor, potentially increasing Npy transcription. In conclusion, we show that BPA-mediated dysregulation of the circadian molecular clock is linked to the deleterious effects of BPA on neuropeptide expression. Furthermore, we describe hypothalamic Bmal1-KO cell lines to study the role of BMAL1 in hypothalamic responses to metabolic, hormonal, and environmental factors.


Subject(s)
ARNTL Transcription Factors/genetics , Benzhydryl Compounds/pharmacology , Endocrine Disruptors/pharmacology , Hypothalamus/drug effects , Neurons/drug effects , Neuropeptide Y/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Period Circadian Proteins/genetics , Phenols/pharmacology , ARNTL Transcription Factors/metabolism , Animals , Circadian Clocks/drug effects , Female , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Neuropeptide Y/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Period Circadian Proteins/metabolism , Promoter Regions, Genetic/drug effects
4.
Neuroendocrinology ; 107(4): 387-399, 2018.
Article in English | MEDLINE | ID: mdl-30352432

ABSTRACT

BACKGROUND AND OBJECTIVES: Elevated levels of saturated fatty acids (SFA) induce a state of neuroinflammation in the hypothalamus. It has been suggested that microglia sense palmitate, a prevalent circulating SFA, and act as mediators of this inflammatory process by communicating with neurons, particularly those involved in appetite regulation. In this study, we examined the inflammatory response to palmitate in immortalized microglial cell lines, BV-2 and IMG, and the subsequent effects on inflammatory gene expression in a model of NPY/AgRP neurons, mHypoE-46. METHODS: The BV-2 cells were treated with 50 µM palmitate for 4 and 24 h, and the transcriptional regulation of markers for inflammation and cellular stress was assessed using an RT2 Profiler PCR Array. Select genes were verified with qRT-PCR. The BV-2 and IMG cells were then co-cultured using 1.0-µm cell culture inserts with an immortalized hypothalamic cell line, mHypoE-46, to investigate potential intercellular communication between microglia and neurons. RESULTS: We found that palmitate increased the mRNA levels of specific inflammatory genes, and a general anti-inflammatory profile was revealed in the microglia cells. The mRNA changes in TNFα at 4 and 24 h in BV-2 cells were abrogated with the toll-like receptor 4 (TLR4) inhibitor, TAK-242, indicating the involvement of TLR4. Co-culture of mHypoE-46 neurons with microglia pre-treated with palmitate resulted in repression of TNFα expression in the hypothalamic neurons. As palmitate significantly increased IL-13 expression in microglia, the effect of this cytokine was tested in mHypoE-46 neurons. The addition of IL-13 to neuronal cultures normalized the palmitate-mediated increase in IL-6 and AgRP expression, suggesting that microglia may protect surrounding neurons, at least in part, through the release of IL-13. CONCLUSIONS: These results suggest a potential anti-inflammatory role of microglia towards the palmitate-induced neuroinflammation, and potentially energy homeostasis, in hypothalamic neurons.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Microglia/drug effects , Neurons/drug effects , Palmitic Acid/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line , Cells, Cultured , Coculture Techniques , Hypothalamus/cytology , Hypothalamus/drug effects , Hypothalamus/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/prevention & control , Lipopolysaccharides/pharmacology , Mice , Microglia/physiology , Neurons/physiology
5.
Am J Physiol Gastrointest Liver Physiol ; 315(2): G206-G219, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29631376

ABSTRACT

Insulin-like growth factor-binding protein-4 (IGFBP-4) is a binding protein that modulates the action of insulin-like growth factor-1 (IGF-1), a growth factor whose presence is required for the intestinotrophic effects of glucagon-like peptide-2 (GLP-2). GLP-2 is a gut hormone that uses both IGF-1 and epidermal growth factor (EGF) as intermediary factors to promote intestinal growth. Therefore, to elucidate the mechanism through which IGFBP-4 regulates IGF-1 activity in the intestine, proliferation assays were conducted using rat intestinal epithelial cells (IEC-6). IGF-1 and EGF synergistically enhanced proliferation, an effect that was dose-dependently decreased by IGFBP-4 ( P < 0.05-0.001) in an IGF-1 receptor (R)- and MEK1/2- but not a phosphatidylinositol 3-kinase-dependent manner ( P > 0.05 for IGFBP-4 effects with IGF-1R and MEK1/2 inhibitors). Intestinal organoids derived from IGFBP-4 knockout mice demonstrated significantly greater Ki-67 expression and an enhanced surface area increase in response to IGF-1 treatment, compared with organoids from control mice ( P < 0.05-0.01). GLP-2 is also known to increase the mucosal expression of IGFBP-4 mRNA. To investigate whether this occurs through the actions of its intermediaries, IGF-1 and EGF, inducible intestinal epithelial-IGF-1R knockout and control mice were treated for 10 days with and without the pan-ErbB inhibitor, CI-1033. However, no differences in mucosal IGFBP-4 mRNA expression were found for any of the treatment groups ( P > 0.05). Consistently, IEC-6 cells treated with IGF-1 and/or EGF displayed no alteration in IGFBP-4 mRNA or in cellular and secreted IGFBP-4 protein ( P > 0.05). Overall, this study establishes that endogenous IGFBP-4 plays an important role in inhibiting IGF-1-induced intestinal epithelial proliferation and that mucosal IGFBP-4 expression is independent of IGF-1 and EGF. NEW & NOTEWORTHY This study demonstrates, for the first time, the inhibitory role of locally expressed insulin-like growth factor-binding protein-4 (IGFBP-4) on the intestinal proliferative actions of IGF-1 and supports the notion of the synergistic roles of IGF-1 and EGF in promoting intestinal epithelial growth. In turn, intestinal IGFBP-4 expression was not found to be regulated by IGF-1 and/or EGF.


Subject(s)
Glucagon-Like Peptide 2/metabolism , Insulin-Like Growth Factor Binding Protein 4/metabolism , Insulin-Like Growth Factor I/metabolism , Intestinal Mucosa/metabolism , Intestines , Animals , Cell Proliferation/physiology , Epidermal Growth Factor/metabolism , Intestines/cytology , Intestines/growth & development , Intestines/physiology , Mice , Rats
6.
Mol Endocrinol ; 30(4): 402-16, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26930171

ABSTRACT

The arcuate nucleus of the hypothalamus represents a key center for the control of appetite and feeding through the regulation of 2 key neuronal populations, notably agouti-related peptide/neuropeptide Y and proopimelanocortin (POMC)/cocaine- and amphetamine-regulated transcript neurons. Altered regulation of these neuronal networks, in particular the dysfunction of POMC neurons upon high-fat consumption, is a major pathogenic mechanism involved in the development of obesity and type 2 diabetes mellitus. Efforts are underway to preserve the integrity or enhance the functionality of POMC neurons in order to prevent or treat these metabolic diseases. Here, we report for the first time that the nitric oxide (NO(-)) donor, sodium nitroprusside (SNP) mediates anorexigenic actions in both hypothalamic tissue and hypothalamic-derived cell models by mediating the up-regulation of POMC levels. SNP increased POMC mRNA in a dose-dependent manner and enhanced α-melanocortin-secreting hormone production and secretion in mHypoA-POMC/GFP-2 cells. SNP also enhanced insulin-driven POMC expression likely by inhibiting the deacetylase activity of sirtuin 1. Furthermore, SNP enhanced insulin-dependent POMC expression, likely by reducing the transcriptional repression of Foxo1 on the POMC gene. Prolonged SNP exposure prevented the development of insulin resistance. Taken together, the NO(-) donor SNP enhances the anorexigenic potential of POMC neurons by promoting its transcriptional expression independent and in cooperation with insulin. Thus, increasing cellular NO(-) levels represents a hormone-independent method of promoting anorexigenic output from the existing POMC neuronal populations and may be advantageous in the fight against these prevalent disorders.


Subject(s)
Appetite Depressants/pharmacology , Insulin/physiology , Neurons/metabolism , Nitroprusside/pharmacology , Pro-Opiomelanocortin/metabolism , Animals , Arcuate Nucleus of Hypothalamus/cytology , Cell Line , Humans , Male , Mice , Neurons/drug effects , Nitric Oxide/physiology , Pro-Opiomelanocortin/genetics , Transcription, Genetic
7.
Mol Endocrinol ; 30(2): 217-33, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26726951

ABSTRACT

Kisspeptin (Kiss) and G-protein-coupled receptor (Gpr)54 have emerged as key regulators of reproduction. 17ß-estradiol (E2)-mediated regulation of these neurons is nuclei specific, where anteroventral periventricular (AVPV) Kiss neurons are positively regulated by E2, whereas arcuate nucleus (ARC) neurons are inhibited. We have generated immortalized Kiss cell lines from male and female adult-derived murine hypothalamic primary culture, as well as cell lines from microdissected AVPV and ARC from female Kiss-green fluorescent protein (GFP) mice. All exhibit endogenous Kiss-1 expression, estrogen receptors (ER)s (ERα, ERß, and Gpr30), as well as known markers of AVPV Kiss neurons in the mHypoA-50 and mHypoA-Kiss/GFP-4, vs markers of ARC Kiss neurons in the mHypoA-55 and the mHypoA-Kiss/GFP-3 lines. There was an increase in Kiss-1 mRNA expression at 24 hours in the AVPV lines and a repression of Kiss-1 mRNA at 4 hours in the ARC lines. An E2-mediated decrease in ERα mRNA expression at 24 hours in the AVPV cell lines was detected, and a significant decrease in Gpr30, ERα, and ERß mRNA levels at 4 hours in the ARC cell lines was evident. ER agonists and antagonists determined the specific ERs responsible for mediating changes in gene expression. In the AVPV, ERα is required but not ERß or GPR30, vs the ARC Kiss-expressing cell lines that require GPR30, and either ERα and/or ERß. We determined cAMP response element-binding protein 1 was necessary for the down-regulation of Kiss-1 mRNA expression using small interfering RNA knockdown in the ARC cell model. These studies elucidate some of the molecular events involved in the differential E2-mediated regulation of unique and specific Kiss neuronal models.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Estradiol/pharmacology , Gene Expression Regulation/drug effects , Hypothalamus, Anterior/metabolism , Kisspeptins/genetics , Receptors, Estrogen/metabolism , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Binding Sites , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Female , Gene Expression Profiling , Gene Knockdown Techniques , Green Fluorescent Proteins/metabolism , Hypothalamus, Anterior/drug effects , Kisspeptins/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Neurons/drug effects , Neurons/metabolism , Peptides/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Estrogen/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Sex Characteristics , Transcription Factor AP-1/metabolism
8.
J Endocrinol ; 220(1): 13-24, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24134870

ABSTRACT

POMC neurons play a central role in the maintenance of whole-body energy homeostasis. This balance requires proper regulation of POMC neurons by metabolic hormones, such as insulin. However, the heterogeneous cellular population of the intact hypothalamus presents challenges for examining the molecular mechanisms underlying the potent anorexigenic effects of POMC neurons, and there is currently a complete lack of mature POMC neuronal cell models for study. To this end, we have generated novel, immortalized, adult-derived POMC-expressing/α-MSH-secreting cell models, mHypoA-POMC/GFP lines 1-4, representing the fluorescence-activated cell-sorted POMC population from primary POMC-eGFP mouse hypothalamus. The presence of Pomc mRNA in these cell lines was confirmed, and α-MSH was detected via immunofluorescence. α-MSH secretion in the mHypoA-POMC/GFP-1 was found to increase in response to 10  ng/ml ciliary neurotrophic factor (CNTF) or 10  nM insulin as determined by enzyme immunoassay. Further experiments using the mHypoA-POMC/GFP-1 cell line revealed that 10  ng/ml CNTF increases Pomc mRNA at 1 and 2  h after treatment, whereas insulin elicited an increase in Pomc mRNA level and decreases in insulin receptor (Insr (Ir)) mRNA level at 4  h. Furthermore, the activation of IR-mediated downstream second messengers was examined by western blot analysis, following the induction of cellular insulin resistance, which resulted in a loss of insulin-mediated regulation of Pomc and Ir mRNAs. The development of these immortalized neurons will be invaluable for the elucidation of the cellular and molecular mechanisms that underlie POMC neuronal function under normal and perturbed physiological conditions.


Subject(s)
Insulin/pharmacology , Neurons/metabolism , Pro-Opiomelanocortin/genetics , Signal Transduction/drug effects , Animals , Blotting, Western , Cell Line, Transformed , Ciliary Neurotrophic Factor/pharmacology , Drug Resistance , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hypoglycemic Agents/pharmacology , Hypothalamus/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/cytology , Phosphorylation/drug effects , Pro-Opiomelanocortin/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , alpha-MSH/metabolism
9.
Mol Cell Endocrinol ; 382(1): 262-270, 2014 Jan 25.
Article in English | MEDLINE | ID: mdl-24145125

ABSTRACT

Glucose-sensing neurons play a role in energy homeostasis, yet how orexigenic neurons sense glucose remains unclear. As models of glucose-inhibited (GI) neurons, mHypoE-29/1 and mHypoA-NPY/GFP cells express the essential orexigenic neuropeptide AgRP and glucose sensing machinery. Exposure to increasing concentrations of glucose or the glucose analog 2-deoxyglucose (2-DG) results in a decrease in AgRP mRNA levels. Taste receptor, Tas1R2 mRNA expression was reduced by glucose, whereas 2-DG reduced Tas1R3 mRNA levels. Increasing glucose concentrations elicited a rise in Akt and neuronal nitric oxide synthase (nNOS) phosphorylation, CaMKKß levels, and a reduction of AMP-kinase alpha phosphorylation. Inhibitors of NOS and the cystic fibrosis transmembrane conductance regulator (CFTR) prevented a decrease in AgRP secretion with glucose, suggesting a pivotal role for nNOS and the CFTR in glucose-sensing. These models possess the hallmark characteristics of GI neurons, and can be used to disentangle the mechanisms by which orexigenic neurons sense glucose.


Subject(s)
Agouti-Related Protein/biosynthesis , Agouti-Related Protein/metabolism , Glucose/pharmacology , Hypothalamus/cytology , Hypothalamus/metabolism , Models, Biological , Adenylate Kinase/metabolism , Agouti-Related Protein/genetics , Animals , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Cell Line , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Deoxyglucose/pharmacology , Down-Regulation/drug effects , Down-Regulation/genetics , Gemfibrozil/pharmacology , Green Fluorescent Proteins/metabolism , Hypothalamus/drug effects , Hypothalamus/embryology , Mice , Neurons/drug effects , Neurons/metabolism , Neuropeptide Y , Nitric Oxide Synthase Type I/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pyruvic Acid/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
10.
Mol Cell Endocrinol ; 377(1-2): 65-74, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23835445

ABSTRACT

Glucose regulates energy homeostasis and reproductive function within the hypothalamus. The underlying mechanisms responsible for glucose regulation of GnRH gene transcription were investigated using a novel murine immortalized, adult-derived hypothalamic cell line, mHypoA-GnRH/GFP. Analysis of GnRH mRNA synthesis and secretion following agonist treatment demonstrated that the mHypoA-GnRH/GFP cell line is a representative model of in vivo GnRH neurons. c-fos mRNA levels, following glucose exposure, indicated that these neurons were responsive to low (0.5mM) and high (5mM) glucose, and high glucose stimulated GnRH mRNA transcription in a metabolism-dependent manner. Glucose inhibited AMPK activity, and was linked to the downstream stimulation of GnRH mRNA levels. The effect was confirmed with an AMPK antagonist, Compound C. Collectively, these findings demonstrate that glucose can directly regulate GnRH transcription, while implicating the AMPK pathway as an essential mediator of nutritional signaling in a novel GnRH neuronal cell model.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aging/metabolism , Glucose/pharmacology , Gonadotropin-Releasing Hormone/metabolism , Green Fluorescent Proteins/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Cell Line , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation/drug effects , Gonadotropin-Releasing Hormone/genetics , Mice , Models, Biological , Neurons , Nitric Oxide/metabolism , Peptides/metabolism , Phosphorylation/drug effects , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Second Messenger Systems , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/pharmacology
11.
Behav Brain Res ; 230(1): 288-90, 2012 Apr 21.
Article in English | MEDLINE | ID: mdl-22366268

ABSTRACT

The expression of passive avoidance (PA) learning in rats displays a daily or circadian rhythm in that optimal performance is displayed when the time of testing matches the time of training. Lesions of the suprachiasmatic nucleus (SCN) were later shown to abolish this rhythm. Using golden hamsters, we have since demonstrated similar rhythms of performance in a conditioned place avoidance (CPA) task but unlike the PA results in rats, the rhythmic expression of CPA was maintained in arrhythmic hamsters with lesions of the SCN. We determined whether PA performance in hamsters is dependent on the SCN (as in the rat) or independent (as in the hamster CPA). Performance on the PA task was rhythmic in intact control animals with optimal performance occurring when training and testing time matched and significantly diminished at both 6h before and 6h after training time. SCN-lesions, verified by the loss of behavioral circadian rhythms, had no effect on the rhythmic expression. Therefore, time of day modulation of PA performance in the hamster does not depend on the SCN circadian clock.


Subject(s)
Avoidance Learning/physiology , Chronobiology Disorders/physiopathology , Conditioning, Operant/physiology , Motor Activity/physiology , Suprachiasmatic Nucleus/physiology , Analysis of Variance , Animals , Chronobiology Disorders/pathology , Cricetinae , Disease Models, Animal , Male , Reaction Time/physiology , Suprachiasmatic Nucleus/injuries
12.
PLoS One ; 6(11): e27799, 2011.
Article in English | MEDLINE | ID: mdl-22132145

ABSTRACT

The pituitary is a complex endocrine tissue composed of a number of unique cell types distinguished by the expression and secretion of specific hormones, which in turn control critical components of overall physiology. The basic function of these cells is understood; however, the molecular events involved in their hormonal regulation are not yet fully defined. While previously established cell lines have provided much insight into these regulatory mechanisms, the availability of representative cell lines from each cell lineage is limited, and currently none are derived from adult pituitary. We have therefore used retroviral transfer of SV40 T-antigen to mass immortalize primary pituitary cell culture from an adult mouse. We have generated 19 mixed cell cultures that contain cells from pituitary cell lineages, as determined by RT-PCR analysis and immunocytochemistry for specific hormones. Some lines expressed markers associated with multipotent adult progenitor cells or transit-amplifying cells, including SOX2, nestin, S100, and SOX9. The progenitor lines were exposed to an adenylate cyclase activator, forskolin, over 7 days and were induced to differentiate to a more mature gonadotrope cell, expressing significant levels of α-subunit, LHß, and FSHß mRNAs. Additionally, clonal populations of differentiated gonadotropes were exposed to 30 nM gonadotropin-releasing hormone and responded appropriately with a significant increase in α-subunit and LHß transcription. Further, exposure of the lines to a pulse paradigm of GnRH, in combination with 17ß-estradiol and dexamethasone, significantly increased GnRH receptor mRNA levels. This array of adult-derived pituitary cell models will be valuable for both studies of progenitor cell characteristics and modulation, and the molecular analysis of individual pituitary cell lineages.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation/drug effects , Cyclic AMP/pharmacology , Gonadotrophs/cytology , Pituitary Gland/cytology , SOXB1 Transcription Factors/metabolism , Stem Cells/cytology , Aging/drug effects , Aging/metabolism , Animals , Biomarkers/metabolism , Cell Line, Transformed , Colforsin/pharmacology , Culture Media, Serum-Free , Follicle Stimulating Hormone, beta Subunit/genetics , Follicle Stimulating Hormone, beta Subunit/metabolism , Gene Expression Regulation/drug effects , Glycoprotein Hormones, alpha Subunit/genetics , Glycoprotein Hormones, alpha Subunit/metabolism , Gonadotrophs/drug effects , Gonadotrophs/metabolism , Luteinizing Hormone/genetics , Luteinizing Hormone/metabolism , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , SOXB1 Transcription Factors/genetics , Stem Cells/drug effects , Stem Cells/metabolism
13.
Endocrinology ; 152(11): 4138-47, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21914774

ABSTRACT

Evidence shows that neuropeptide Y (NPY) neurons are involved in mediating the anorexigenic action of leptin via neuronal circuits in the hypothalamus. However, studies have produced limited data on the cellular processes involved and whether hypothalamic NPY neurons are susceptible to cellular leptin resistance. To investigate the direct regulation of NPY secretion by leptin, we used novel NPY-synthesizing, immortalized mHypoA-NPY/green fluorescent protein and mHypoA-59 hypothalamic cell lines derived from adult hypothalamic primary cultures. We report that leptin treatment significantly suppressed NPY secretion in the cells by approximately 20%. We found a decrease in c-fos expression upon leptin exposure, indicating deactivation or hyperpolarization of the neurons. Protein analysis indicated that leptin inhibits AMP-activated protein kinase (AMPK) activity and activates acetyl-coenzyme A carboxylase in NPY neurons, supporting the hypothesis of an AMPK-dependent mechanism. Inhibiting both AMPK with Compound C or phosphatidylinositol 3 kinase (PI3K) with 2-(4-morpholinyl)-8-phenyl-1(4H)-1-benzopyran-4-one hydrochloride prevented the leptin-mediated decrease in NPY secretion, indicating both AMPK- and PI3K-mediated mechanisms. Further, NPY secretion was stimulated by 30% by the AMPK activator, aminoimidazole carboxamide ribonucleotide. Importantly, prolonged leptin exposure in the mHypoA-NPY/green fluorescent protein cells prevented leptin-induced changes in AMPK phosphorylation and suppression of NPY secretion, indicating that NPY neurons are susceptible to leptin resistance. Our studies indicate that AMPK and PI3K pathways are involved in leptin action in NPY neurons and that leptin resistance blocks the feedback response likely required to maintain energy homeostasis.


Subject(s)
Hypothalamus/metabolism , Leptin/metabolism , Neurons/metabolism , Neuropeptide Y/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line , Mice , Phosphorylation
14.
FASEB J ; 23(12): 4256-65, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19703933

ABSTRACT

The distinct lack of cell lines derived from the adult brain is evident. Ciliary neurotrophic factor (CNTF) triggers neurogenesis in primary culture from adult mouse hypothalamus, as detected by bromodeoxyuridine and Ki67 immunostaining. Using SV-40 T-antigen, we immortalized dividing neurons and generated clonal cell lines expressing neuropeptides and receptors involved in neuroendocrine function. We hypothesized that proglucagon-derived peptides may be the mechanistic downstream effectors of CNTF due to documented neuroprotective and proliferative effects. Indeed, proglucagon gene expression was induced by CNTF, and exposure of primary cells to glucagon-like peptide-1 receptor (GLP-1) agonist, exendin-4, induced cell proliferation. Intracerebroventricular injection of CNTF into adult mice caused increased expression of proglucagon peptide in the hypothalamus. Using a specific GLP-1-receptor antagonist, we found that neurogenesis was significantly attenuated and primary culture from GLP-1-receptor-knockout mice lacked CNTF-mediated neuronal proliferation, thus linking the induction of neurogenesis in the hypothalamus to GLP-1-receptor signaling.


Subject(s)
Ciliary Neurotrophic Factor/pharmacology , Glucagon-Like Peptide 1/metabolism , Hypothalamus/cytology , Neurogenesis/physiology , Neurons/cytology , Animals , Cell Line , Cell Proliferation , Ciliary Neurotrophic Factor/metabolism , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Proglucagon/genetics , Proglucagon/metabolism , Signal Transduction
15.
Am J Physiol Regul Integr Comp Physiol ; 295(5): R1529-38, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18768761

ABSTRACT

The circadian system in mammals is a hierarchy of oscillators throughout the organism that are coordinated by the circadian clock in the hypothalamic suprachiasmatic nucleus. Peripheral clocks act to integrate time-of-day information from neural or hormonal signals, regulating gene expression, and, subsequently, organ physiology. However, the mechanisms by which the central clock communicates with peripheral oscillators are not understood and are likely tissue specific. In this study, we establish a mouse vascular cell model suitable for investigations of these mechanisms at a molecular level. Using the immortalized vascular smooth muscle cell line Movas-1, we determined that these cells express the circadian clock machinery with robust rhythms in mRNA expression over a 36-h period after serum shock synchronization. Furthermore, norepinephrine and forskolin were able to synchronize circadian rhythms in bmal1. With synchronization, we observed cycling of specific genes, including the tissue inhibitor of metalloproteinase 1 and 3 (timp1, timp3), collagen 3a1 (col3a1), transgelin 1 (sm22alpha), and calponin 1 (cnn1). Diurnal expression of these genes was also found in vivo in mouse aortic tissue, using microarray and real-time RT-PCR analysis. Both of these revealed ultradian rhythms in genes similar to the cycling observed in Movas-1 in vitro. These findings highlight the cyclical nature of structurally important genes in the vasculature that is similar both in vivo and in vitro. This study establishes the Movas-1 cells as a novel cell model from which to further investigate the molecular mechanisms of clock regulation in the vasculature.


Subject(s)
Circadian Rhythm/genetics , Circadian Rhythm/physiology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Animals , Aorta/cytology , Aorta/physiology , Cell Line , Colforsin/pharmacology , Computational Biology , Culture Media , DNA, Complementary/biosynthesis , DNA, Complementary/isolation & purification , Gene Expression Regulation/physiology , Male , Mice , Mice, Inbred C57BL , Norepinephrine/pharmacology , Oligonucleotide Array Sequence Analysis , RNA/biosynthesis , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Vasoconstrictor Agents/pharmacology
16.
J Mol Endocrinol ; 41(3): 117-24, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18550896

ABSTRACT

Neuroendocrine peptides express biologic activity relevant to the cardiovascular system, including regulating heart rate and blood pressure, though little is known about the mechanisms involved. Here, we investigated neuroendocrine gene expression underlying diurnal physiology of the heart. We first used microarray and RT-PCR analysis and demonstrate the simultaneous expression of neuroendocrine genes in normal murine heart, including POMC, GnRH, neuropeptide Y, leptin receptor, GH-releasing hormone, cocaine- and amphetamine-regulated transcript, proglucagon, and galanin. We examined diurnal gene expression profiles, with cosinar bioinformatics to evaluate statistically significant rhythms. The POMC gene exhibits a day/night, circadian or diurnal, pattern of expression in heart, and we postulated that this may be important to cardiac growth and renewal. POMC diurnal gene rhythmicity is altered in pressure-overload cardiac hypertrophy, when compared with control heart, and levels increased at the dark-to-light transition times. These findings are also consistent with the proposal that neuropeptides mediate adverse remodeling processes, such as occur in pathologic hypertrophy. To investigate cellular responses, we screened three cell lines representing fibroblasts, cardiac myocytes, and vascular smooth muscle cells (NIH3T3, heart line 1, and mouse vascular smooth muscle cell line 1 (Movas-1) respectively). POMC mRNA expression is the most notable in Movas-1 cells and, furthermore, exhibits rhythmicity with culture synchronization. Taken together, these results highlight the diverse neuroendocrine mRNA expression profiles in cardiovasculature, and provide a novel model vascular culture system to research the role these neuropeptides play in organ health, integrity, and disease.


Subject(s)
Cardiomegaly/genetics , Cardiomegaly/physiopathology , Circadian Rhythm/physiology , Gene Expression Regulation , Myocardium/metabolism , Neurosecretory Systems/metabolism , Pro-Opiomelanocortin/genetics , Animals , Aorta/pathology , Blood Pressure , Constriction, Pathologic/genetics , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/metabolism , NIH 3T3 Cells , Neuropeptides/genetics , Neuropeptides/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
17.
Oecologia ; 143(4): 566-77, 2005 May.
Article in English | MEDLINE | ID: mdl-15791425

ABSTRACT

Herbivore-induced plants responses can affect the preference and performance of herbivores and their natural enemies. These responses may vary depending on the identity and number of herbivore species feeding on the plant so that when herbivores from different guilds feed on plants, the interactions between plants, herbivores, and natural enemies may be disrupted. Tomato plants were damaged either by the caterpillar Spodoptera exigua, or the aphid Macrosiphum euphorbiae, or damaged by both herbivores, or undamaged controls. We measured the preference and performance of S. exigua and its parasitoid Cotesia marginiventris, and activity of proteinase inhibitors (PI) as an indicator of induced resistance. Compared to undamaged plants, caterpillar damage reduced the number of eggs laid by S. exigua adults, reduced growth, consumption, and survival of larval S. exigua and C. marginiventris, and increased activity of PIs 43%; but did not increase attraction of C. marginiventris. While pupal mass of S. exigua was not affected, the pupal mass of C. marginiventris decreased on caterpillar-damaged plants compared to controls. In contrast, plants damaged by aphids were preferred for oviposition by S. exigua, and had increased larval consumption and survival, compared to controls. Aphid feeding did not affect the preference or performance of C. marginiventris, or PI activity, compared to controls. While oviposition was deterred on caterpillar-damaged plants, plants damaged by both herbivores received the same amount of oviposition as controls. The attraction of C. marginiventris to plants damaged by caterpillars and aphids was increased compared to controls. However, plants damaged by both herbivores had similar PI activity, larval growth and survival of S. exigua and C. marginiventris, as plants singly damaged by caterpillars. Overall, the preference component for both the herbivore and parasitoid was more strongly affected by damage due to multiple herbivores than the performance component.


Subject(s)
Aphids/physiology , Feeding Behavior/physiology , Solanum lycopersicum/physiology , Spodoptera/growth & development , Wasps/growth & development , Analysis of Variance , Animals , Female , Host-Parasite Interactions , Larva/growth & development , Oviposition/physiology , Protease Inhibitors/metabolism , Spodoptera/parasitology
18.
Neurobiol Learn Mem ; 81(3): 217-20, 2004 May.
Article in English | MEDLINE | ID: mdl-15082023

ABSTRACT

In golden hamsters, the expression of a conditioned place preference (CPP) or avoidance (CPA) is regulated in a circadian pattern such that the preference and avoidance are exhibited strongly at the circadian time of prior training, but not at other circadian times. In the rat, reports are conflicting regarding whether time of day learning is evident. We investigated whether this conflict arises because different strains of rat have been used. In this experiment, Long Evans and Wistar rats were trained at a specific circadian time to discriminate between a context paired with food reward and an unpaired context. Animals were then tested for preference at the same or a different circadian time. Long Evans rats showed preference for the paired context at both times tested, whereas Wistar rats showed preference only when training and testing times matched. The results show that time of day learning can be generalized to rats using the Wistar strain.


Subject(s)
Association Learning/physiology , Circadian Rhythm/physiology , Conditioning, Classical/physiology , Recognition, Psychology/physiology , Reinforcement, Psychology , Animals , Housing, Animal , Male , Rats , Rats, Long-Evans , Rats, Wistar , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...