Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Endocrinology ; 158(7): 2391-2405, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28368536

ABSTRACT

This study aims at investigating the epigenetic landscape of cardiomyocytes exposed to elevated glucose levels. High glucose (30 mM) for 72 hours determined some epigenetic changes in mouse HL-1 and rat differentiated H9C2 cardiomyocytes including upregulation of class I and III histone deacetylase protein levels and activity, inhibition of histone acetylase p300 activity, increase in histone H3 lysine 27 trimethylation, and reduction in H3 lysine 9 acetylation. Gene expression analysis focused on cardiotoxicity revealed that high glucose induced markers associated with tissue damage, fibrosis, and cardiac remodeling such as Nexilin (NEXN), versican, cyclic adenosine 5'-monophosphate-responsive element modulator (CREM), and adrenoceptor α2A (ADRA2). Notably, the transcription factor CREM was found to be important in the regulation of cardiotoxicity-associated genes as assessed by specific small interfering RNA and chromatin immunoprecipitation experiments. In CD1 mice, made hyperglycemic by streptozotoicin (STZ) injection, cardiac structural alterations were evident at 6 months after STZ treatment and were associated with a significant increase of H3 lysine 27 trimethylation and reduction of H3 lysine 9 acetylation. Consistently, NEXN, CREM, and ADRA2 expression was significantly induced at the RNA and protein levels. Confocal microscopy analysis of NEXN localization showed this protein irregularly distributed along the sarcomeres in the heart of hyperglycemic mice. This evidence suggested a structural alteration of cardiac Z-disk with potential consequences on contractility. In conclusion, high glucose may alter the epigenetic landscape of cardiac cells. Sildenafil, restoring guanosine 3', 5'-cyclic monophosphate levels, counteracted the increase of CREM and NEXN, providing a protective effect in the presence of hyperglycemia.


Subject(s)
Cardiotoxicity/genetics , Cyclic AMP Response Element Modulator/physiology , Glucose/adverse effects , Glucose/metabolism , Hyperglycemia/metabolism , Hyperglycemia/pathology , Myocytes, Cardiac/metabolism , Animals , Cardiotoxicity/metabolism , Cardiotoxicity/pathology , Cells, Cultured , Cyclic AMP Response Element Modulator/genetics , Disease Models, Animal , Embryo, Mammalian , Epigenesis, Genetic/drug effects , Female , Hyperglycemia/genetics , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Rats , Time Factors
2.
Hepatology ; 61(3): 990-1002, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25330465

ABSTRACT

UNLABELLED: Molecular factors driving immune-mediated inflammation in the liver are incompletely understood. The transcription factor, cyclic adenosine monophosphate-responsive element modulator alpha (CREMα) can endorse differentiation of T lymphocytes toward T-helper (Th)17 cells, thereby promoting autoimmunity in systemic lupus erythematosus or lung inflammation. To investigate the role of CREMα in liver disease, we subjected transgenic (Tg) mice overexpressing CREMα under control of the CD2 promoter (cremtg mice), which restrains expression mainly to lymphocytes (T, natural killer [NK], and NKT cells), to acute and chronic liver injury models. Already in steady state, Tg CREMα overexpression broadly reduced hepatic immune cell numbers by decreasing their viability, but did not affect immune cell migration or the fibrogenic response to chronic liver injury. Strikingly, cremtg mice developed more severe immune-mediated hepatitis with a higher mortality rate, compared to wild-type (wt) mice, upon concanavalin A (ConA) administration. Unlike in T cells from spleen, CREMα overexpression did not induce a predominant Th17 response in intrahepatic T cells, given that hepatic cremtg CD4+ T cells expressed less interleukin (IL)-17 than wt T cells. Reconstitution of Rag1-/- mice with Crem-/- T cells did not ameliorate ConA hepatitis. Overexpression of CREMα did not influence NK and NKT-cell effector functions either. Interestingly, a subset of monocytic myeloid-derived suppressor cells (MDSCs) also expressed CD2 and CREMα. Cremtg MDSCs isolated from liver expressed reduced inducible nitric oxide synthase and arginase 1 and displayed a reduced T-cell suppressive activity. The adoptive transfer of wt MDSCs was capable of reducing the fulminant immune-mediated liver damage in cremtg mice to wt level. CONCLUSION: These results suggest compartmental differences of T cell activation pathways between liver and other organs in autoimmunity and define a functional role of CREMα in hepatic monocytic MDSCs for the pathogenesis of immune-mediated liver disease.


Subject(s)
Cyclic AMP Response Element Modulator/physiology , Hepatitis, Autoimmune/etiology , Liver/immunology , Myeloid Cells/physiology , Adoptive Transfer , Animals , CD2 Antigens/analysis , Cell Differentiation , Concanavalin A , Liver Cirrhosis, Experimental/etiology , Mice , Mice, Transgenic , T-Lymphocytes/immunology , Th17 Cells/physiology
3.
Thyroid ; 24(1): 78-87, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23819433

ABSTRACT

BACKGROUND: Although adenosine monophosphate activated protein kinase (AMPK) plays a crucial role in energy metabolism, a direct effect of AMPK modulation on thyroid function has only recently been reported, and much of its function in the thyroid is currently unknown. The aim of this study was to investigate the mechanism of AMPK modulation in iodide uptake. Furthermore, we wanted to investigate the potential of the AMPK inhibitor compound C as an enhancer of iodide uptake by thyrocytes. METHODS: The in vitro and in vivo effects of AMPK modulation on sodium-iodide symporter (NIS) protein levels and iodide uptake were examined in follicular rat thyroid cell-line cells and C57Bl6/J mice. Activation of AMPK by metformin resulted in a strong reduction of iodide uptake (up to sixfold with 5 mM metformin after 96 h) and NIS protein levels in vitro, whereas AMPK inhibition by compound C not only stimulated iodide uptake but also enhanced NIS protein levels both in vitro (up to sevenfold with 1 µM compound C after 96 h) and in vivo (1.5-fold after daily injections with 20 mg/kg for 4 days). We investigated the regulation of NIS expression by AMPK using a range of promoter constructs consisting of either the NIS promoter or isolated CRE (cAMP response element) and NF-κB elements, which are present within the NIS promoter. RESULTS: Metformin reduced NIS promoter activity (0.6-fold of control), whereas compound C stimulated its activity (3.4-fold) after 4 days. This largely coincides with CRE activation (0.6- and 3.0-fold). These experiments show that AMPK exerts its effects on iodide uptake, at least partly, through the CRE element in the NIS promoter. Furthermore, we have used AMPK-alpha1 knockout mice to determine the long-term effects of AMPK inhibition without chemical compounds. These mice have a less active thyroid, as shown by reduced colloid volume and reduced responsiveness to thyrotropin. CONCLUSION: NIS expression and iodine uptake in thyrocytes can be modulated by metformin and compound C. These compounds exert their effect by modulation of AMPK, which, in turn, regulates the activation of the CRE element in the NIS promoter. Overall, this suggests that the use of AMPK modulating compounds may be useful for the enhancement of iodide uptake by thyrocytes, which could be useful for the treatment of thyroid cancer patients with radioactive iodine.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Iodides/metabolism , Metformin/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Symporters/biosynthesis , Thyroid Gland/metabolism , Animals , Cell Line , Cyclic AMP Response Element Modulator/physiology , Male , Mice , Mice, Inbred C57BL , Rats , Symporters/drug effects , Thyroid Gland/drug effects
4.
Eur J Immunol ; 42(6): 1375-84, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22678893

ABSTRACT

Elevated levels of intracellular cyclic adenosine monophosphate (cAMP) in naturally occurring T regulatory (nTreg) cells play a key role in nTreg-cell-mediated suppression. Upon contact with nTreg cells, cAMP is transferred from nTreg cells into activated target CD4(+) T cells and/or antigen-presenting cells (APCs) via gap junctions to suppress CD4(+) T-cell function. cAMP facilitates the expression and nuclear function of a potent transcriptional inhibitor, inducible cAMP early repressor (ICER), resulting in ICER-mediated suppression of interleukin-2 (IL-2). Furthermore, ICER inhibits transcription of nuclear factor of activated T cell c1/α (NFATc1/α) and forms inhibitory complexes with preexisting NFATc1/c2, thereby inhibiting NFAT-driven transcription, including that of IL-2. In addition to its suppressive effects mediated via ICER, cAMP can also modulate the levels of surface-expressed cytotoxic T lymphocyte antigen-4 (CTLA-4) and its cognate B7 ligands on conventional CD4(+) T cells and/or APCs, fine-tuning suppression. These cAMP-driven nTreg-cell suppression mechanisms are the focus of this review.


Subject(s)
Cyclic AMP/physiology , T-Lymphocytes, Regulatory/physiology , Animals , Antigen-Presenting Cells/physiology , B7-1 Antigen/physiology , CD28 Antigens/physiology , CTLA-4 Antigen/physiology , Cyclic AMP Response Element Modulator/physiology , Forkhead Transcription Factors/physiology , Homeostasis , Humans , Interleukin-2/antagonists & inhibitors , Interleukin-2/biosynthesis , NFATC Transcription Factors/metabolism , Signal Transduction
5.
PLoS One ; 7(2): e31798, 2012.
Article in English | MEDLINE | ID: mdl-22384077

ABSTRACT

The essential role of the Crem gene in normal sperm development is widely accepted and is confirmed by azoospermia in male mice lacking the Crem gene. The exact number of genes affected by Crem absence is not known, however a large difference has been observed recently between the estimated number of differentially expressed genes found in Crem knock-out (KO) mice compared to the number of gene loci bound by CREM. We therefore re-examined global gene expression in male mice lacking the Crem gene using whole genome transcriptome analysis with Affymetrix microarrays and compared the lists of differentially expressed genes from Crem-/- mice to a dataset of genes where binding of CREM was determined by Chip-seq. We determined the global effect of CREM on spermatogenesis as well as distinguished between primary and secondary effects of the CREM absence. We demonstrated that the absence of Crem deregulates over 4700 genes in KO testis. Among them are 101 genes associated with spermatogenesis 41 of which are bound by CREM and are deregulated in Crem KO testis. Absence of several of these genes in mouse models has proven their importance for normal spermatogenesis and male fertility. Our study showed that the absence of Crem plays a more important role on different aspects of spermatogenesis as estimated previously, with its impact ranging from apoptosis induction to deregulation of major circadian clock genes, steroidogenesis and the cell-cell junction dynamics. Several new genes important for normal spermatogenesis and fertility are down-regulated in KO testis and are therefore possible novel targets of CREM.


Subject(s)
Cyclic AMP Response Element Modulator/physiology , Gene Expression Regulation , Testis/metabolism , Animals , Apoptosis , Biological Transport , Cyclic AMP Response Element Modulator/biosynthesis , Fertilization , Gene Expression Profiling , Homozygote , Male , Melatonin/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction/methods , Spermatids/metabolism , Spermatogenesis , Spermatozoa/metabolism
6.
Front Neuroendocrinol ; 33(1): 67-84, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21871477

ABSTRACT

Corticotropin releasing hormone (CRH) is essential for stress adaptation by mediating hypothalamic-pituitary-adrenal (HPA) axis, behavioral and autonomic responses to stress. Activation of CRH neurons depends on neural afferents from the brain stem and limbic system, leading to sequential CRH release and synthesis. CRH transcription is required to restore mRNA and peptide levels, but termination of the response is essential to prevent pathology associated with chronic elevations of CRH and HPA axis activity. Inhibitory feedback mediated by glucocorticoids and intracellular production of the repressor, Inducible Cyclic AMP Early Repressor (ICER), limit the magnitude and duration of CRH neuronal activation. Induction of CRH transcription is mediated by the cyclic AMP/protein kinase A/cyclic AMP responsive element binding protein (CREB)-dependent pathways, and requires cyclic AMP-dependent nuclear translocation of the CREB co-activator, Transducer of Regulated CREB activity (TORC). This article reviews current knowledge on the mechanisms regulating CRH neuron activity.


Subject(s)
Corticotropin-Releasing Hormone/physiology , Neurons/physiology , Animals , Base Sequence , Corticotropin-Releasing Hormone/biosynthesis , Cyclic AMP/physiology , Cyclic AMP Response Element Modulator/physiology , Cyclic AMP Response Element-Binding Protein/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Epigenesis, Genetic , Feedback , Glucocorticoids/physiology , Humans , Mice , Molecular Sequence Data , Paraventricular Hypothalamic Nucleus/growth & development , Rats , Signal Transduction/physiology , Stress, Physiological/physiology , Transcription Factors/physiology , Vasopressins/biosynthesis
8.
Article in English | MEDLINE | ID: mdl-21555408

ABSTRACT

Spermatogenesis is a complex and ordered differentiation process in which the spermatogonial stem cell population gives rise to primary spermatocytes that undergo two successive meiotic divisions followed by a major biochemical and structural reorganization of the haploid cells to generate mature elongate spermatids. The transcriptional regulatory programs that orchestrate this process have been intensively studied in model organisms such as Drosophila melanogaster and mouse. Genetic and biochemical approaches have identified the factors involved and revealed mechanisms of action that are unique to male germ cells. In a well-studied example, cofactors and pathways distinct from those used in somatic tissues mediate the action of CREM in male germ cells. But perhaps the most striking feature concerns the paralogs of somatically expressed transcription factors and of components of the general transcription machinery that act in distinct regulatory mechanisms in both Drosophila and murine spermatogenesis.


Subject(s)
Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Germ Cells/metabolism , Animals , Cyclic AMP Response Element Modulator/genetics , Cyclic AMP Response Element Modulator/physiology , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Haploidy , Male , Meiosis/genetics , Mice , Models, Genetic , Spermatogenesis/genetics , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/physiology
9.
Diabetologia ; 54(9): 2337-46, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21547497

ABSTRACT

AIMS/HYPOTHESIS: Pro-atherogenic and pro-oxidant, oxidised LDL trigger adverse effects on pancreatic beta cells, possibly contributing to diabetes progression. Because oxidised LDL diminish the expression of genes regulated by the inducible cAMP early repressor (ICER), we investigated the involvement of this transcription factor and of oxidative stress in beta cell failure elicited by oxidised LDL. METHODS: Isolated human and rat islets, and insulin-secreting cells were cultured with human native or oxidised LDL or with hydrogen peroxide. The expression of genes was determined by quantitative real-time PCR and western blotting. Insulin secretion was monitored by EIA kit. Cell apoptosis was determined by scoring cells displaying pycnotic nuclei. RESULTS: Exposure of beta cell lines and islets to oxidised LDL, but not to native LDL raised the abundance of ICER. Induction of this repressor by the modified LDL compromised the expression of important beta cell genes, including insulin and anti-apoptotic islet brain 1, as well as of genes coding for key components of the secretory machinery. This led to hampering of insulin production and secretion, and of cell survival. Silencing of this transcription factor by RNA interference restored the expression of its target genes and alleviated beta cell dysfunction and death triggered by oxidised LDL. Induction of ICER was stimulated by oxidative stress, whereas antioxidant treatment with N-acetylcysteine or HDL prevented the rise of ICER elicited by oxidised LDL and restored beta cell functions. CONCLUSIONS/INTERPRETATION: Induction of ICER links oxidative stress to beta cell failure caused by oxidised LDL and can be effectively abrogated by antioxidant treatment.


Subject(s)
Cyclic AMP Response Element Modulator/physiology , Insulin-Secreting Cells/physiology , Islets of Langerhans/physiopathology , Oxidative Stress/physiology , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Cells, Cultured , Cyclic AMP Response Element Modulator/drug effects , Cyclic AMP Response Element Modulator/genetics , Humans , Hydrogen Peroxide/pharmacology , Insulin/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Lipoproteins, LDL/pharmacology , Male , Models, Animal , Oxidative Stress/drug effects , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley
10.
J Neurochem ; 115(3): 563-73, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20367754

ABSTRACT

Repeated exposure to opiates leads to cellular and molecular changes and behavioral alterations reflecting a state of dependence. In noradrenergic neurons, cyclic AMP (cAMP)-dependent pathways are activated during opiate withdrawal, but their contribution to the activity of locus coeruleus noradrenergic neurons and behavioral manifestations remains controversial. Here, we test whether the cAMP-dependent transcription factors cAMP responsive element binding protein (CREB) and cAMP-responsive element modulator (CREM) in noradrenergic neurons control the cellular markers and the physical signs of morphine withdrawal in mice. Using the Cre/loxP system we ablated the Creb1 gene in noradrenergic neurons. To avoid adaptive effects because of compensatory up-regulation of CREM, we crossed the conditional Creb1 mutant mice with a Crem-/- line. We found that the enhanced expression of tyrosine hydroxylase normally observed during withdrawal was attenuated in CREB/CREM mutants. Moreover, the withdrawal-associated cellular hyperactivity and c-fos expression was blunted. In contrast, naloxone-precipitated withdrawal signs, such as jumping, paw tremor, tremor and mastication were preserved. We conclude by a specific genetic approach that the withdrawal-associated hyperexcitability of noradrenergic neurons depends on CREB/CREM activity in these neurons, but does not mediate several behavioral signs of morphine withdrawal.


Subject(s)
Cyclic AMP Response Element Modulator/genetics , Cyclic AMP Response Element Modulator/physiology , Cyclic AMP Response Element-Binding Protein/physiology , Locus Coeruleus/physiology , Morphine Dependence/psychology , Norepinephrine/physiology , Substance Withdrawal Syndrome/psychology , Sympathetic Nervous System/physiology , Animals , Brain/anatomy & histology , Cell Survival/genetics , Chromatography, High Pressure Liquid , Chronic Disease , Cyclic AMP Response Element-Binding Protein/genetics , Electrochemistry , Electrophysiology , Female , Genotype , In Situ Hybridization , Locus Coeruleus/cytology , Male , Mice , Mice, Knockout , Morphine/adverse effects , Morphine Dependence/physiopathology , Narcotics/adverse effects , Substance Withdrawal Syndrome/physiopathology , Sympathetic Nervous System/cytology , Transcription Factors/physiology
11.
J Clin Invest ; 119(7): 1940-51, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19603549

ABSTRACT

A trial fibrillation (AF), the most common human cardiac arrhythmia, is associated with abnormal intracellular Ca2+ handling. Diastolic Ca2+ release from the sarcoplasmic reticulum via "leaky" ryanodine receptors (RyR2s) is hypothesized to contribute to arrhythmogenesis in AF, but the molecular mechanisms are incompletely understood. Here, we have shown that mice with a genetic gain-of-function defect in Ryr2 (which we termed Ryr2R176Q/+ mice) did not exhibit spontaneous AF but that rapid atrial pacing unmasked an increased vulnerability to AF in these mice compared with wild-type mice. Rapid atrial pacing resulted in increased Ca2+/calmodulin-dependent protein kinase II (CaMKII) phosphorylation of RyR2, while both pharmacologic and genetic inhibition of CaMKII prevented AF inducibility in Ryr2R176Q/+ mice. This result suggests that AF requires both an arrhythmogenic substrate (e.g., RyR2 mutation) and enhanced CaMKII activity. Increased CaMKII phosphorylation of RyR2 was observed in atrial biopsies from mice with atrial enlargement and spontaneous AF, goats with lone AF, and patients with chronic AF. Genetic inhibition of CaMKII phosphorylation of RyR2 in Ryr2S2814A knockin mice reduced AF inducibility in a vagotonic AF model. Together, these findings suggest that increased RyR2-dependent Ca2+ leakage due to enhanced CaMKII activity is an important downstream effect of CaMKII in individuals susceptible to AF induction.


Subject(s)
Atrial Fibrillation/etiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Calcium/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Atrial Fibrillation/metabolism , Atrial Fibrillation/prevention & control , Cardiac Pacing, Artificial , Cyclic AMP Response Element Modulator/physiology , Electrocardiography , Goats , Heart Atria/metabolism , Heart Atria/pathology , Humans , Mice , Mice, Transgenic , Peptides/physiology , Phosphorylation , Ryanodine Receptor Calcium Release Channel/physiology
12.
Mol Neurobiol ; 40(1): 73-86, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19434522

ABSTRACT

The inducible cAMP early repressor (ICER) is an endogenous repressor of cAMP-responsive element (CRE)-mediated gene transcription and belongs to the CRE-binding protein (CREB)/CRE modulator (CREM)/activating transcription factor 1 (ATF-1) gene family. ICER plays an important role in regulating the neuroendocrine system and the circadian rhythm. Other aspects of ICER function have recently attracted heightened attention. Being a natural inducible CREB antagonist, and more broadly, an inducible repressor of CRE-mediated gene transcription, ICER regulates long-lasting plastic changes that occur in the brain in response to incoming stimulation. This review will bring together data on ICER and its functions in the brain, with a special emphasis on recent findings highlighting the involvement of ICER in the regulation of long-term plasticity underlying learning and memory.


Subject(s)
Brain/physiology , Cyclic AMP Response Element Modulator/physiology , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation , Humans , Learning/physiology , Memory/physiology , Neuronal Plasticity
13.
J Neurosci ; 29(2): 328-33, 2009 Jan 14.
Article in English | MEDLINE | ID: mdl-19144833

ABSTRACT

The family of CREB (cAMP response element-binding protein) transcription factors are involved in a variety of biological processes including the development and plasticity of the nervous system. In the maturing and adult brain, CREB genes are required for activity-dependent processes, including synaptogenesis, refinement of connections and long-term potentiation. Here, we use CREB1(Nescre)CREM(-/-) (cAMP-responsive element modulator) mutants to investigate the role of these genes in stimulus-independent patterns of neural activity at early stages. We show that lack of CREB/CREM genes specifically in neural tissue leads to increased synaptogenesis and to a dramatic increase in the levels of spontaneous network activity at embryonic stages. Thus, the functions of CREB/CREM genes in neural activity differ in distinct periods of neural development.


Subject(s)
Cyclic AMP Response Element Modulator/physiology , Cyclic AMP Response Element-Binding Protein/physiology , Neural Pathways/physiology , Neurons/physiology , Synapses/genetics , Age Factors , Animals , Brain/cytology , Brain/embryology , Brain/metabolism , Calcium/metabolism , Cyclic AMP Response Element Modulator/deficiency , Cyclic AMP Response Element-Binding Protein/deficiency , Embryo, Mammalian , In Vitro Techniques , Mice , Mice, Knockout , Neural Pathways/ultrastructure , Neurons/ultrastructure , Synapses/ultrastructure
14.
J Neurosci ; 28(25): 6459-72, 2008 Jun 18.
Article in English | MEDLINE | ID: mdl-18562617

ABSTRACT

Long-lasting neuronal plasticity as well as long-term memory (LTM) requires de novo synthesis of proteins through dynamic regulation of gene expression. cAMP-responsive element (CRE)-mediated gene transcription occurs in an activity-dependent manner and plays a pivotal role in neuronal plasticity and LTM in a variety of species. To study the physiological role of inducible cAMP early repressor (ICER), a CRE-mediated gene transcription repressor, in neuronal plasticity and LTM, we generated two types of ICER mutant mice: ICER-overexpressing (OE) mice and ICER-specific knock-out (KO) mice. Both ICER-OE and ICER-KO mice show no apparent abnormalities in their development and reproduction. A comprehensive battery of behavioral tests revealed no robust changes in locomotor activity, sensory and motor functions, and emotional responses in the mutant mice. However, long-term conditioned fear memory was attenuated in ICER-OE mice and enhanced in ICER-KO mice without concurrent changes in short-term fear memory. Furthermore, ICER-OE mice exhibited retardation of kindling development, whereas ICER-KO mice exhibited acceleration of kindling. These results strongly suggest that ICER negatively regulates the neuronal processes required for long-term fear memory and neuronal plasticity underlying kindling epileptogenesis, possibly through suppression of CRE-mediated gene transcription.


Subject(s)
Cyclic AMP Response Element Modulator/physiology , Epilepsy/metabolism , Fear/physiology , Kindling, Neurologic/metabolism , Memory/physiology , Repressor Proteins/physiology , Animals , Cyclic AMP Response Element Modulator/genetics , Epilepsy/genetics , Female , Kindling, Neurologic/genetics , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neural Inhibition/physiology
15.
Endocrinology ; 149(5): 2592-606, 2008 May.
Article in English | MEDLINE | ID: mdl-18202121

ABSTRACT

The lack of Na(+)/I(-) symporter (NIS) gene expression in some thyroid cancer patients has been a major hurdle that limits the efficacy of standard radioactive iodide therapy. The molecular mechanism that contributes to low NIS expression is not well understood. Activated NIS gene expression is stimulated by thyroid-stimulating hormone-mediated cAMP/protein kinase A signaling through a NIS upstream enhancer (NUE). The cAMP pathway is also stimulated by forskolin. In the current work, we studied the mechanism of transcriptional activation of NIS in normal thyroid cells and thyroid cancer cells. We identified the cAMP response element modulator (CREM) activator as a new component of the transcription complex that is important for NIS gene expression. The CREM complex is seen in the normal thyroid cells and BRAF (V600E) thyroid cancer cells (BHP 17-10) but is missing in rearranged in transformation/papillary thyroid carcinoma-1 rearrangement thyroid cancer cells (BHP 2-7). This complex is believed to be responsible for the loss of NUE activity and reduced NIS expression in the BHP 2-7 cell line. In BHP 2-7 cells, forskolin stimulated the thyroid-specific transcription factor Pax 8, but CREM activator mRNA did not increase, and this produced a small increase in NUE activity. Ectopic expression of CREM activator enhanced activity of the NUE, indicating that CREM is an essential regulator of NIS gene expression.


Subject(s)
Cyclic AMP Response Element Modulator/physiology , Enhancer Elements, Genetic , Symporters/genetics , Animals , Carcinoma, Papillary/genetics , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP Response Element Modulator/metabolism , Enhancer Elements, Genetic/drug effects , Gene Expression Regulation, Neoplastic , Humans , PAX8 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Protein Binding , Proteins/metabolism , Proto-Oncogene Proteins B-raf/genetics , Rats , Thyroid Neoplasms/genetics , Transcriptional Activation
16.
J Neurosci Res ; 86(1): 61-70, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17722060

ABSTRACT

Programmed cell death involving gene regulation and de novo protein synthesis is a major component of both normal development and a number of disease conditions. Hence, knowledge of its mechanisms, especially transcription factors, that regulate expression of the genes involved in neurodegenerative disorders is of great importance. cAMP-responsive element-binding protein (CREB) has repeatedly been implicated in the neuronal survival. In the present study we showed that inducible cAMP early repressor (ICER), an endogenous CREB antagonist, is expressed during both excitotoxic and spontaneous neuronal cell death in organotypic hippocampal slice cultures in vitro. Furthermore, overexpression of ICER via an adenoviral vector evoked neuronal cell loss in such cultures. The time course of ICER-dependent cell death was hippocampal subdivision specific, with dentate gyrus neurons dying mostly 3-7 days after the adenovector infection, followed by CA3, where neuronal death peaked after 7 days, and then CA1, where most neuronal death occurred after 7-14 days. These results underscore the usefulness of the organotypic cultures for studies of neurodegeneration and point to neuronal loss having a multifaceted nature in a complex cellular environment.


Subject(s)
Cyclic AMP Response Element Modulator/physiology , Gene Expression Regulation/physiology , Hippocampus/cytology , Neurons/physiology , Animals , Animals, Newborn , Cell Death/drug effects , Cell Death/physiology , Excitatory Amino Acid Agonists/toxicity , Gene Expression Regulation/drug effects , Genetic Vectors/physiology , Microtubule-Associated Proteins/metabolism , N-Methylaspartate/toxicity , Neurons/drug effects , Organ Culture Techniques , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Wistar , Time Factors , Transfection/methods
17.
Eur J Neurosci ; 26(4): 810-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17666079

ABSTRACT

Traumatic brain injury (TBI) results in significant hippocampal pathology and hippocampal-dependent memory loss, both of which are alleviated by hypothermia treatment. To elucidate the molecular mechanisms regulated by hypothermia after TBI, rats underwent moderate parasagittal fluid-percussion brain injury. Brain temperature was maintained at normothermic or hypothermic temperatures for 30 min prior and up to 4 h after TBI. The ipsilateral hippocampus was assayed with Western blotting. We found that hypothermia potentiated extracellular signal-regulated kinase 1/2 (ERK1/2) activation and its downstream effectors, p90 ribosomal S6 kinase (p90RSK) and the transcription factor cAMP response element-binding protein. Phosphorylation of another p90RSK substrate, Bad, also increased with hypothermia after TBI. ERK1/2 regulates mRNA translation through phosphorylation of mitogen-activated protein kinase-interacting kinase 1 (Mnk1) and the translation factor eukaryotic initiation factor 4E (eIF4E). Hypothermia also potentiated the phosphorylation of both Mnk1 and eIF4E. Augmentation of ERK1/2 activation and its downstream signalling components may be one molecular mechanism that hypothermia treatment elicits to improve functional outcome after TBI.


Subject(s)
Brain Injuries/enzymology , Brain Injuries/therapy , Hypothermia, Induced , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Animals , Blotting, Western , Brain Injuries/psychology , Cyclic AMP Response Element Modulator/physiology , Enzyme Activation/physiology , Eukaryotic Initiation Factor-4E/biosynthesis , Eukaryotic Initiation Factor-4E/genetics , Hippocampus/physiology , Immunohistochemistry , Learning/physiology , Learning Disabilities/etiology , Learning Disabilities/psychology , Male , Microscopy, Confocal , Phosphorylation , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases, 90-kDa/physiology , Signal Transduction/physiology
18.
Horm Metab Res ; 39(8): 560-6, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17712720

ABSTRACT

The MC2-Receptor (melanocortin 2 receptor, MC2-R) is a Gs-protein coupled receptor that is upregulated by its own ligand ACTH and by forskolin. The mechanisms regulating MC2-R expression are still unclear. We therefore investigated the role of the stimulatory transcription factors CREB and CREM and the inhibitory factor ICER for regulation of human MC2-R expression. We cotransfected mouse adrenocortical Y1 cells with luciferase reporter gene vectors containing full length and deleted human MC2-R promoter constructs with expression plasmids for CREB, CREBS133A, CREMtau, CREMtauS117A, or ICER. Direct protein-DNA interaction was investigated by EMSA. Wild type CREB did not significantly affect promoter activity due to high endogenous CREB activity. However, CREBS133A decreased forskolin stimulated MC2-R promoter activity by 48+/-5% (mean+/-SEM) while unstimulated values remained unchanged. CREMtau moderately increased basal and forskolin stimulated luciferase activity in a dose-dependent manner (maximum effect 252+/-24% and 186+/-13% VS. control vector, respectively). While this effect required the full length promoter, cAMP stimulation was retained in shorter constructs. ICER reduced basal luciferase activity in Y1 cells by 17+/-28%, but completely abolished forskolin stimulation. Although 5'-deletion constructs mapped the minimum promoter region required for ICER effect to the shortest -64/+40 construct, direct protein DNA interaction in this promoter region could not be identified by EMSA. Moreover, mutation of the SF-1 binding sites, which retained ICER dependent inhibition, excluded SF-1 to be required for this effect. We conclude from these data that transcription factors of the CREB/CREM/ATF family have a moderate effect on human MC2-R promoter activity, but seem to play a minor role in transmitting stimulation of the cAMP pathway to increased MC2-R expression.


Subject(s)
Adrenal Cortex/metabolism , Cyclic AMP Response Element Modulator/physiology , Cyclic AMP Response Element-Binding Protein/physiology , Receptor, Melanocortin, Type 2/genetics , Animals , Cell Line , Colforsin/pharmacology , Cyclic AMP/pharmacology , Gene Expression Regulation , Humans , Mice , Promoter Regions, Genetic/drug effects , Transfection
19.
J Immunol ; 179(1): 607-15, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17579082

ABSTRACT

Communication between the nervous and immune systems involves the release of neuropeptides, such as calcitonin gene-related peptide (CGRP), from sensory nerves during inflammation. CGRP may inhibit the activities of both innate and adaptive immune cells, but the molecular pathways underlying this function are largely unknown. In this study, we identify CGRP as a potent inhibitor of TLR-stimulated production of inflammatory mediators, such as TNF-alpha and CCL4, by murine dendritic cells. Inhibition of TLR responses was independent of IL-10 and did not involve perturbation of canonical TLR signaling, including activation of MAPK and NF-kappaB. Instead, the inhibitory activity of CGRP was mediated by the cAMP/protein kinase A pathway leading to rapid up-regulation of the transcriptional repressor, inducible cAMP early repressor (ICER). Ectopically expressed ICER directly repressed the LPS-stimulated activity of a synthetic Tnf promoter, as well as TNF-alpha protein production driven by the endogenous promoter. Inhibition of dendritic cell gene expression by CGRP was associated with the presence of a composite cAMP response element/kappaB promoter element. In a murine model of endotoxemia, CGRP markedly attenuated serum TNF-alpha levels, and this effect was associated with the up-regulation of ICER. Together, these results establish a novel pathway for the negative regulation of TLR responses through the nervous system that critically involves induction of the transcriptional repressor ICER by the neuropeptide CGRP.


Subject(s)
Calcitonin Gene-Related Peptide/physiology , Cyclic AMP Response Element Modulator/physiology , Down-Regulation/immunology , Repressor Proteins/physiology , Toll-Like Receptors/antagonists & inhibitors , Toll-Like Receptors/physiology , Animals , Calcitonin Gene-Related Peptide/administration & dosage , Calcitonin Gene-Related Peptide/metabolism , Cell Line , Cyclic AMP/biosynthesis , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endotoxemia/immunology , Female , Humans , Lipopolysaccharides/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Calcitonin Gene-Related Peptide/immunology , Receptors, Calcitonin Gene-Related Peptide/metabolism , Transcription, Genetic , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics
20.
Stress ; 10(2): 153-61, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17514584

ABSTRACT

Corticotropin releasing factor (CRF) coordinates behavioral, autonomic and hormonal responses to stress. Activation of the hypothalamic pituitary adrenal (HPA) axis with stimulation of CRF and vasopressin (VP) release from hypothalamic parvocellular neurons, and consequent secretion of ACTH from the anterior pituitary and glucocorticoid from the adrenal cortex, is the major endocrine response to stress. Current evidence indicates that the main regulator of ACTH secretion in acute and chronic conditions is CRF, in spite of the fact that the selective increases in expression of parvocellular VP and pituitary VP V1b receptors observed during prolonged activation of the HPA axis have suggested that VP becomes the predominant regulator. Following CRF release, activation of CRF transcription is required to restore mRNA and peptide levels, but termination of the response is essential to prevent pathology associated with chronic elevation of CRF and glucocorticoid production. While glucocorticoid feedback plays an important role in regulating CRF expression, the relative importance of direct transcriptional repression of the CRF gene by glucocorticoids in the overall feedback mechanism is not clear. In addition to glucocorticoids, intracellular feedback mechanisms in the CRF neuron, involving induction of repressor forms of cAMP response element modulator (CREM) limit CRF transcriptional responses by competing with the positive regulator, phospho-CREB. Rapid repression of CRF transcription following stress-induced activation is likely to contribute to limiting the stress response and to preventing disorders associated with excessive CRF production.


Subject(s)
Corticotropin-Releasing Hormone/biosynthesis , Stress, Physiological/physiopathology , Adrenal Glands/physiology , Animals , Cyclic AMP Response Element Modulator/physiology , Cyclic AMP Response Element-Binding Protein/physiology , Gene Expression Regulation/drug effects , Glucocorticoids/physiology , Paraventricular Hypothalamic Nucleus/physiology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...