hosted by
publicationslist.org
    

patricia parnet


patricia.parnet@univ-nantes.fr

Journal articles

2010
Bérengère Coupé, Valérie Amarger, Isabelle Grit, Alexandre Benani, Patricia Parnet (2010)  Nutritional programming affects hypothalamic organization and early response to leptin.   Endocrinology 151: 2. 702-713 Feb  
Abstract: Nutritional programming, taking place in utero or early after birth, is closely linked with metabolic and appetite disorders in adulthood. Following the hypothesis that nutritional programming impacts hypothalamic neuronal organization, we report on discrepancies of multiple molecular and cellular early events that take place in the hypothalamus of rats submitted to intrauterine growth restriction (IUGR). Expression screening performed on hypothalami from IUGR rats at birth and at postnatal d 12 identified changes in gene expression of neurodevelopmental process (cell differentiation and cytoskeleton organization). Additionally, a slight reduction of agouti-related protein and a strong reduction of alpha-MSH-immunoreactive efferent fibers were demonstrated in the paraventricular nucleus of IUGR rats. Rapid catch-up growth of IUGR rats, 5 d after birth, had a positive effect on neurodevelopmental factors and on neuronal projections emanating from the arcuate nucleus. The molecular and cellular anomalies detected in IUGR rats can be related to the reduced and delayed plasma leptin surge from d 0-16 when compared with control and IUGR rats with catch-up growth. However, the ability of leptin to activate intracellular signaling in arcuate nucleus neurons was not reduced in IUGR rats. Other mechanism such as epigenetic regulation of the major appetite-regulating neuropeptides genes was analyzed in parallel with their mRNA expression during postnatal development. This study reveals the importance of an early catch-up growth that reduces abnormal organization of hypothalamic pathways involved in energy homeostasis, whereas protein restriction, maintained during postnatal development leads to an important immaturity of the hypothalamus.
Notes:
2009
Bérengère Coupé, Isabelle Grit, Dominique Darmaun, Patricia Parnet (2009)  The timing of "catch-up growth" affects metabolism and appetite regulation in male rats born with intrauterine growth restriction.   Am J Physiol Regul Integr Comp Physiol 297: 3. R813-R824 Sep  
Abstract: Epidemiological studies demonstrated a relationship between low birth weight mainly caused by intrauterine growth restriction (IUGR) and adult metabolic disorders. The concept of metabolic programming centers on the idea that nutritional and hormonal status during the key period of development determines the long-term control of energy balance by programming future feeding behavior and energy expenditure. The present study examined the consequence of early or late "catch-up growth" after IUGR on feeding behavior and metabolic cues of male offspring of rat dams exposed to protein restriction during gestation and/or lactation. Our results suggest that early catch-up growth may be favorable for fasting metabolic parameters at weaning, as no differences were observed on plasma leptin, triglyceride, glucose, and insulin levels compared with controls. In contrast, if pups remained malnourished until weaning, low insulin concentration was detected and was accompanied by hyperphagia associated with a large increase in hypothalamic NPY and AgRP mRNA expression. At adult age, on a regular chow diet, only the meal structure was modified by fetal programming. The two IUGR groups demonstrated a reduced meal duration that enhanced the speed of food ingestion and consequently increased the rest period associated to the satiety state without changes in the hypothalamic expression of appetite neuropeptides. Our findings demonstrate that in IUGR, regardless of postnatal growth magnitude, metabolic programming occurred in utero and was responsible for both feeding behavior alteration and postprandial higher insulin level in adults. Additionally, catch-up growth immediately after early malnutrition could be a key point for the programming of postprandial hyperleptinemia.
Notes:
2005
Vincent Marty, Chantal Médina, Chantal Combe, Patricia Parnet, Thierry Amédée (2005)  ATP binding cassette transporter ABC1 is required for the release of interleukin-1beta by P2X7-stimulated and lipopolysaccharide-primed mouse Schwann cells.   Glia 49: 4. 511-519 Mar  
Abstract: Schwann cells are best known as myelinating glial cells of the peripheral nervous system, but they also participate actively in the sphere of immunity by producing pro-inflammatory cytokines, such as interleukin-1beta (IL-1beta). In a previous study, we demonstrated that posttranslational processing of IL-1beta by immune-challenged Schwann cells required the P2X7 receptor. Remarkably, the release of IL-1beta was not associated with cell death, indicating the involvement of an active mechanism. ATP binding cassette (ABC) transporters are known to transport leaderless secretory proteins, such as IL-1beta; therefore, we investigated whether such transporters were at work in Schwann cells. Mouse Schwann cells expressed ABC1 transporter mRNA and displayed the functional protein. Glybenclamide and diisothiocyanato-stilbene-disulfonic acid (DIDS), two blockers of chloride fluxes that drive the export activity of ABC1 transporters, inhibited IL-1beta release without altering its intracellular processing. Enhancing chloride efflux potentiated the release of IL-1beta, while decreasing it led to a strong reduction in its release. Because the stimulation of the P2X7 receptor also activates a chloride conductance, we investigated the possibility of a sole anionic pathway mobilized by the P2X7 receptor and ABC1. Glybenclamide and DIDS had no significant effects on the P2X7-activated chloride current suggesting therefore the existence of two different pathways. In summary, ABC1 transporters are required for the release of IL-1beta by mouse Schwann cells. Being associated together with chloride conductance, P2X7 receptors and ABC1 transporters delineate a subtle and complex regulation of IL-1beta production in mammalian Schwann cells. Furthermore, ABC1 transporters could be a target of therapeutic interest for regulating IL-1beta activity in neuroinflammation disorders.
Notes:
Agnès Nadjar, Viviane Tridon, Michael J May, Sankar Ghosh, Robert Dantzer, Thierry Amédée, Patricia Parnet (2005)  NFkappaB activates in vivo the synthesis of inducible Cox-2 in the brain.   J Cereb Blood Flow Metab 25: 8. 1047-1059 Aug  
Abstract: Interleukin-1beta (IL-1beta) induces cyclooxygenase-2 (Cox-2) expression in many of its cellular targets resulting in production and release of prostaglandins. Although IL-1beta-induced Cox-2 expression most likely requires activation of nuclear transcription factor kappa B (NFkappaB) pathway, this has never been formally demonstrated in vivo. We tested this using a specific inhibitor of NFkappaB activation, the NEMO binding domain (NBD) peptide, that has been shown previously to be effective in various in vivo models of acute inflammation. Incubation of rat glioma cells with the NBD peptide blocked IL-1beta-induced NFkappaB nuclear translocation. Furthermore, after injection of a biotinylated version of the NBD peptide into the lateral ventricle of the brain, we found that it readily diffused to its potential cellular targets in vivo. To test the effects of the peptide on NFkappaB activation and Cox-2 expression in the brain, we injected it intracerebroventricularly (36 microg/rat) into rats before intraperitoneal injection of IL-1beta (60 microg/kg). Treatment with NBD peptide completely abolished IL-1beta-induced NFkappaB activation and Cox-2 synthesis in microvasculature. In contrast, the peptide had no effect on constitutive neuronal Cox-2. These findings strongly support the hypothesis that IL-1beta-induced NFkappaB activation plays a major role in transmission of immune signals from the periphery to the brain.
Notes:
Agnès Nadjar, Rose-Marie Bluthé, Michael J May, Robert Dantzer, Patricia Parnet (2005)  Inactivation of the cerebral NFkappaB pathway inhibits interleukin-1beta-induced sickness behavior and c-Fos expression in various brain nuclei.   Neuropsychopharmacology 30: 8. 1492-1499 Aug  
Abstract: The behavioral effects of peripherally administered interleukin-1beta (IL-1beta) are mediated by the production of cytokines and other proinflammatory mediators at the level of the blood-brain interface and by activation of neural pathway. To assess whether this action is mediated by NFkappaB activation, rats were injected into the lateral ventricle of the brain with a specific inhibitor of NFkappaB activation, the NEMO Binding Domain (NBD) peptide that has been shown previously to abolish completely IL-1beta-induced NFkappaB activation and Cox-2 synthesis in the brain microvasculature. NFkappaB pathway inactivation significantly blocked the behavioral effects of intraperitoneally administered IL-1beta in the form of social withdrawal and decreased food intake, and dramatically reduced IL-1beta-induced c-Fos expression in various brain regions as paraventricular nucleus, supraoptic nucleus, and lateral part of the central amygdala. These findings strongly support the hypothesis that IL-1beta-induced NFkappaB activation at the blood-brain interface is a crucial step in the transmission of immune signals from the periphery to the brain that underlies further events responsible of sickness behavior.
Notes:
A Nadjar, C Combe, P Busquet, R Dantzer, P Parnet (2005)  Signaling pathways of interleukin-1 actions in the brain: anatomical distribution of phospho-ERK1/2 in the brain of rat treated systemically with interleukin-1beta.   Neuroscience 134: 3. 921-932  
Abstract: Interleukin-1beta is released at the periphery during infection and acts on the nervous system to induce fever, neuroendocrine activation, and behavioral changes. These effects are mediated by brain type I IL-1 receptors. In vitro studies have shown the ability of interleukin-1beta to activate mitogen-activated protein kinase signaling pathways including p38, c-Jun N-terminal kinase and extracellular signal-regulated protein kinase 1 and 2 (ERK1/2). In contrast to other mitogen-activated protein kinases, little is known about ERK1/2 activation in the rat brain in response to interleukin-1beta. The aim of the present study was therefore to investigate spatial and temporal activation of ERK1/2 in the rat brain after peripheral administration of interleukin-1beta using immunohistochemistry to detect the phosphorylated form of the kinase. In non-stimulated conditions, phosphorylated ERK1/2 immunoreactivity was observed in neurons throughout the brain. Administration of interleukin-1beta (60 microg/kg, i.p.) induced the phosphorylation of ERK1/2 in areas at the interface between brain and blood or cerebrospinal fluid: meninges, circumventricular organs, endothelial like cells of the blood vessels, and in brain nuclei involved in behavioral depression, fever and neuroendocrine activation: paraventricular nucleus of the hypothalamus, supraoptic nucleus, central amygdala and arcuate nucleus. Double labeling of phosphorylated ERK1/2 and cell markers revealed the expression of phosphorylated ERK1/2 in neurons, astrocytes and microglia. Since phosphorylated ERK1/2 was found in structures in which type I IL-1 receptor has already been identified as well as in structures lacking this receptor, activation of ERK1/2 is likely to occur in response to both direct and indirect action of interleukin-1beta on its target cells.
Notes:
2003
Aurore Colomar, Vincent Marty, Chantal Médina, Chantal Combe, Patricia Parnet, Thierry Amédée (2003)  Maturation and release of interleukin-1beta by lipopolysaccharide-primed mouse Schwann cells require the stimulation of P2X7 receptors.   J Biol Chem 278: 33. 30732-30740 Aug  
Abstract: The P2X7 receptor, mainly expressed by immune cells, is a ionotropic receptor activated by high concentration of extracellular ATP. It is involved in several processes relevant to immunomodulation and inflammation. Among these processes, the production of extracellular interleukin-1beta (IL-1beta), a pro-inflammatory cytokine, plays a major role in the activation of the cytokine network. We have investigated the role of P2X7 receptor and of an associated calcium-activated potassium conductance (BK channels) in IL-1beta maturation and releasing processes by Schwann cells. Lipopolysaccharide-primed Schwann cells synthesized large amounts of pro-IL-1beta but did not release detectable amounts of pro or mature IL-1beta. ATP on its own had no effect on the synthesis of pro-IL-1beta, but a co-treatment with lipopolysaccharide and ATP led to the maturation and the release of IL-1beta by Schwann cells. Both mechanisms were blocked by oxidized ATP. IL-1beta-converting enzyme (ICE), the caspase responsible for the maturation of pro-IL-1beta in IL-1beta, was activated by P2X7 receptor stimulation. The specific inhibition of ICE by the caspase inhibitor Ac-Tyr-Val-Ala-Asp-aldehyde blocked the maturation of IL-1beta. In searching for a link between the P2X7 receptor and the activation of ICE, we found that enhancing potassium efflux from Schwann cells upregulated the production of IL-1beta, while strongly reducing potassium efflux led to opposite effects. Blocking BK channels actually modulated IL-1beta release. Taken together, these results show that P2X7 receptor stimulation and associated BK channels, through the activation of ICE, leads to the maturation and the release of IL-1beta by immune-challenged Schwann cells.
Notes:
P Parnet, F Pousset, S Laye (2003)  NF kappa B activation in mouse pituitary: comparison of response to interleukin-1 beta and lipopolysaccharide.   J Neuroendocrinol 15: 3. 304-314 Mar  
Abstract: The mouse anterior pituitary contains both types of interleukin (IL)-1 receptors, IL-1 receptor type I (IL-1RI) and IL-1 receptor type II (IL-1RII). These receptors are expressed mainly on somatotroph cells. In the present study, the ability of the mouse pituitary to respond in vivo to IL-1 or to lipopolysaccharide (LPS) was demonstrated by measuring, with an electrophoretic mobility shift assay, the presence of an active NF kappa B complex in cell nuclei from pituitaries of mice injected intraperitoneally with recombinant rat-IL-1 beta or LPS. Using immunohistochemistry with an antibody directed against the p65 NF kappa B subunit, a rapid and transient NF kappa B response to LPS was observed. This response was present predominantly in the nuclei of glial fibrillary acidic protein (GFAP)-positive cells and F4/80-labelled cells of the posterior and the anterior pituitary 15 min after stimulation and became faint after 2 h. In comparison, the early and strong NF kappa B response to IL-1 beta treatment was localized into somatotroph cells, GFAP positive cells and F4/80-labelled cells of the posterior and anterior pituitary. Activation of NF kappa B in response to IL-1 beta was no longer apparent in IL-1RI knockout mice, confirming that this receptor is essential for the transduction of IL-1 signal in the pituitary, but remained after LPS treatment. In addition, we investigated the effect of IL-1 on target genes by measuring the mRNA and proteins synthesis of growth hormone (GH), IL-6 and IL-1ra in the pituitary and the plasma. IL-1 beta was shown to induce a rapid and strong synthesis of IL-6 and IL-1ra in the pituitary but failed to regulate GH contents or release. These data suggest that the pituitary is able to respond to a systemic infection via cytokine-mediated responses transduced by IL-1.
Notes:
Agnès Nadjar, Chantal Combe, Sophie Layé, Viviane Tridon, Robert Dantzer, Thierry Amédée, Patricia Parnet (2003)  Nuclear factor kappaB nuclear translocation as a crucial marker of brain response to interleukin-1. A study in rat and interleukin-1 type I deficient mouse.   J Neurochem 87: 4. 1024-1036 Nov  
Abstract: The signalling pathways that mediate early central effects of interleukin-1 (IL-1) during the acute phase reaction have been poorly elucidated. Interaction of IL-1beta to its specific receptor interleukin-1 receptor type I (IL-1RI) leads to nuclear factor kappa B (NuFkappaB) nuclear translocation and a robust transcriptional activation of inhibitor of kappa B alpha (IkappaBalpha) within the rat brain. Indeed, we demonstrated that IL-1RI expressed in blood brain barrier (BBB) cells and in circumventricular organs (CVOs) is crucial for p65-NFkappaB translocation induced by peripheral injection of IL-1beta. Moreover, it has been previously shown that monitoring IkappaBalpha mRNA synthesis is an effective tool to investigate the activity of the transcription factor NFkappaB into the CNS. However in the present study we observed time-related and cell-type differences between IkappaBalpha mRNA synthesis and p65-NFkappaB translocation. This indicates that the expression of IkappaBalpha mRNA does not strictly parallel p65-NFkappaB nuclear translocation, suggesting that these markers are not interchangeable to investigate NFkappaB activity but must be studied together. Thus, we hypothesize that IL-1beta reached the brain across the CVOs that lack a BBB and endothelial cells all over the brain and interacted with its receptors to induce NFkappaB translocation. The study of the consequences of the impairment of NFkappaB pathway activation in in vivo experimentation should bring important clues about the precise role of this transcription factor.
Notes:
Aurore Colomar, Vincent Marty, Chantal Combe, Chantal Médina, Patricia Parnet, Thierry Amédée (2003)  The immune status of Schwann cells: what is the role of the P2X7 receptor?   J Soc Biol 197: 2. 113-122  
Abstract: The peripheral nervous system (PNS) displays structural barriers and a lack of lymphatic drainage which strongly limit the access of molecules and cells from the immune system. In addition, the PNS has the ability to set up some specific mechanisms of immune protection to limit the pathogenicity of inflammation processes following insults by pathogens or inflammatory autoimmune diseases like the Guillain-Barré syndrome. Schwann cells are among the most prominent cells which can display immune capabilities in the PNS. Numerous in vitro studies have shown that Schwann cells were indeed able to display a large repertoire of properties, ranging from the participation to antigen presentation, to secretion of pro- and anti-inflammatory cytokines, chemokines and neurotrophic factors. In vivo studies have confirmed the immune capabilities of Schwann cells. The aim of this review is to present how Schwann cells can participate to the initiation, the regulation and the termination of the immune response in the light of the recent discovery of the Schwann cell expression of purinergic P2X7 receptors.
Notes:
2002
Patricia Parnet, Keith W Kelley, Rose Marie Bluthé, Robert Dantzer (2002)  Expression and regulation of interleukin-1 receptors in the brain. Role in cytokines-induced sickness behavior.   J Neuroimmunol 125: 1-2. 5-14 Apr  
Abstract: Sickness behavior refers to the coordinated set of behavior changes that develop in sick individuals during the course of an infection. At the molecular level, these changes are due to the effects of proinflammatory cytokines as interleukin-1 on the brain. The purpose of this article is not to review the entire field of cytokines and behavior, but rather to address the role of interleukin-1 receptors (IL-1Rs) in sickness behavior. We briefly describe the notion of sickness behavior and present the distribution of IL-1Rs in the central nervous system of the human, mouse and rat. We then bring arguments in favor of the functionality of the various subtypes of receptors and evaluate the nature of the signaling pathways activated by brain IL-1Rs to initiate central modifications leading to symptoms of sickness. Finally, modulation of IL-1 action on its receptor by various opposing factors including glucocorticoids and anti-inflammatory cytokines is discussed.
Notes:
Jan Pieter Konsman, Patricia Parnet, Robert Dantzer (2002)  Cytokine-induced sickness behaviour: mechanisms and implications.   Trends Neurosci 25: 3. 154-159 Mar  
Abstract: Sickness behaviour represents the expression of the adaptive reorganization of the priorities of the host during an infectious episode. This process is triggered by pro-inflammatory cytokines produced by peripheral phagocytic cells in contact with invading micro-organisms. The peripheral immune message is relayed to the brain via a fast neural pathway and a slower humoral pathway, resulting in the expression of pro-inflammatory cytokines in macrophage-like cells and microglia in the brain. The cellular and molecular components of this previously unsuspected system are being progressively identified. These advances are opening new avenues for understanding brain disorders, including depression.
Notes:
2001
F Pousset, S Cremona, R Dantzer, K W Kelley, P Parnet (2001)  Dexamethasone up-regulates type II IL-1 receptor in mouse primary activated astrocytes.   J Neurochem 76: 3. 901-909 Feb  
Abstract: Brain astrocytes play a pivotal role in the brain response to inflammation. They express IL-1 receptors including the type I IL-1 receptor (IL-1RI) that transduces IL-1 signals in cooperation with the IL-1 receptor accessory protein (IL-1RAcP) and the type II IL-1 receptor (IL-1RII) that functions as a decoy receptor. As glucocorticoid receptors are expressed on astrocytes, we hypothesized that glucocorticoids regulate IL-1 receptors expression. IL-1beta-activated mouse primary astrocytes were treated with 10(-6) M dexamethasone, and IL-1 receptors were studied at the mRNA and protein levels. Using RT-PCR, IL-1RI and IL-1RII but not IL-1RAcP mRNAs were found to be up-regulated by dexamethasone in a time-dependent manner. Dexamethasone (Dex), but not progesterone, had no effect on IL-1RI but strongly increased IL-1RII mRNA expression. Binding studies revealed an increase in the number of IL-1RII binding sites under the effect of Dex, but no change in affinity. These findings support the concept that glucocorticoids have important regulatory effect on the response of astrocytes to IL-1.
Notes:
F Pousset, S Cremona, R Dantzer, K W Kelley, P Parnet (2001)  IL-10 and IL-4 regulate type-I and type-II IL-1 receptors expression on IL-1 beta-activated mouse primary astrocytes.   J Neurochem 79: 4. 726-736 Nov  
Abstract: When activated by its ligand, the interleukin receptor type I (IL-1RI) transduces signals in cooperation with the IL-1 receptor accessory protein (IL-1RacP). In contrast, IL-1RII functions as a decoy receptor without participating in IL-1 signalling. Brain astrocytes are cellular targets of IL-1 and play a pivotal role in brain responses to inflammation. The regulation of IL-1 receptors on astrocytes by anti-inflammatory cytokines such as IL-4 and IL-10 has not been studied, despite its importance for understanding the way these cells respond to IL-1. Using RT-PCR, we first showed that the expression of IL-1RI and IL-1RII, but not IL-1RacP, mRNAs are up-regulated by IL-1 beta in a time-dependent manner. Using a radioligand binding technique, we then showed that astrocytes display an equivalent number of IL-1RI and IL-1RII. IL-1 beta decreases the number of IL-1RI binding sites, whereas it increases those of IL-1RII. IL-4 and IL-10 both up-regulate IL-1RII IL-1 beta-induced, but only IL-4 does so for IL-1RI. At the protein level, IL-4 and IL-10 dramatically reverse the ability of IL-1 beta to inhibit expression of IL-1RI but neither affects the ability of IL-1 beta to enhance the number of IL-1RII. Collectively, these results establish the existence of receptor cross-talk between pro- and anti-inflammatory cytokines on a critical type of cell that regulates inflammatory events in the brain.
Notes:
K Palin, F Pousset, D Verrier, R Dantzer, K Kelley, P Parnet, J Lestage (2001)  Characterization of interleukin-1 receptor antagonist isoform expression in the brain of lipopolysaccharide-treated rats.   Neuroscience 103: 1. 161-169  
Abstract: The endogenous interleukin-1 receptor antagonist is the natural inhibitor of the biological effects of interleukin-1 during inflammation. Interleukin-1 receptor antagonist refers to three isoforms: one secreted and two intracellular forms (types I and II). The objective of the present study was to investigate the expression of interleukin-1 receptor antagonist isoforms in the rat brain in vivo in response to an i.p. injection of lipopolysaccharide. The interleukin-1 receptor antagonist was studied at the messenger and protein levels by reverse transcription-polymerase chain reaction and western blot analysis, respectively. Interleukin-1 receptor antagonist messenger RNA was constitutively expressed in the brain and its expression increased in response to lipopolysaccharide. The three interleukin-1 receptor antagonist protein isoforms were up-regulated after lipopolysaccharide treatment in a time-dependent manner. Their relative expression differed according to the isoform and brain region studied. Double immunofluorescence staining revealed interleukin-1 receptor antagonist positive neurons and microglia in hippocampus 24h after lipopolysaccharide stimulation. These results demonstrate for the first time that brain cells are able to produce interleukin-1 receptor antagonist isoforms in response to a peripheral immune challenge with a predominance of the secreted over intracellular forms.
Notes:
2000
S Layé, G Gheusi, S Cremona, C Combe, K Kelley, R Dantzer, P Parnet (2000)  Endogenous brain IL-1 mediates LPS-induced anorexia and hypothalamic cytokine expression.   Am J Physiol Regul Integr Comp Physiol 279: 1. R93-R98 Jul  
Abstract: The present study was designed to determine the role of endogenous brain interleukin (IL)-1 in the anorexic response to lipopolysaccharide (LPS). Intraperitoneal administration of LPS (5-10 microgram/mouse) induced a dramatic, but transient, decrease in food intake, associated with an enhanced expression of proinflammatory cytokine mRNA (IL-1beta, IL-6, and tumor necrosis factor-alpha) in the hypothalamus. This dose of LPS also increased plasma levels of IL-1beta. Intracerebroventricular pretreatment with IL-1 receptor antagonist (4 microgram/mouse) attenuated LPS-induced depression of food intake and totally blocked the LPS-induced enhanced expression of proinflammatory cytokine mRNA measured in the hypothalamus 1 h after treatment. In contrast, LPS-induced increases in plasma levels of IL-1beta were not altered. These findings indicate that endogenous brain IL-1 plays a pivotal role in the development of the hypothalamic cytokine response to a systemic inflammatory stimulus.
Notes:
F Pousset, R Dantzer, K W Kelley, P Parnet (2000)  Interleukin-1 signaling in mouse astrocytes involves Akt: a study with interleukin-4 and IL-10.   Eur Cytokine Netw 11: 3. 427-434 Sep  
Abstract: Although astrocytes are well known to respond to the pro-inflammatory cytokine, interleukin-1 (IL-1), the receptor and post-receptor mechanisms that mediate IL-1 effects in this cell type are complex and need further investigation. Using electrophoretic mobility shift assay (EMSA), we show that IL-1beta-induced NFkappaB activation in primary culture of mouse astrocytes is mediated by the interaction of this cytokine with the IL-1 type I receptor/IL-1 receptor accessory protein complex, as demonstrated by the ability of blocking monoclonal antibodies against these receptors to attenuate NFkappaB activation. In addition to NFkappaB activation, IL-1beta is also able to phosphorylate Akt, as demonstrated by Western blot. The observation that addition of wortmanin, that specifically blocks Akt phosphorylation, also attenuates NFkappaB activation can be interpreted that Akt phosphorylation interacts with IL-1 signaling pathways. Furthermore, anti-inflammatory cytokines such as IL-4 and IL-10 that block IL-1b-induced NFkappaB activation also attenuate IL-1beta-induced Akt phosphorylation, despite the fact that IL-4 and IL-10 in isolation induced Akt phosphorylation. All these findings point to an interaction between Akt and NFkappaB-dependent IL-1 signaling in the primary culture of astrocytes.
Notes:
S Liège, E Moze, K W Kelley, P Parnet, P J Neveu (2000)  Activation of the hypothalamic-pituitary-adrenal axis in IL-1 beta-converting enzyme-deficient mice.   Neuroimmunomodulation 7: 4. 189-194  
Abstract: Interleukin-1beta (IL-1beta) plays a key role in immune, behavioral and neuroendocrine responses to inflammation or infection. IL-1beta could also be involved in the response of the hypothalamic-pituitary-adrenal (HPA) axis during stress. Mature IL-1beta derives from a 31-kD precursor (pro-IL-1beta) that is processed by IL-1beta-converting enzyme (ICE). Mice in which the ICE gene has been nullated by homologous recombination were used to investigate the role of IL-1beta in the HPA axis response. Plasma levels of corticosterone and adrenocorticotropic hormone (ACTH) in response to an intraperitoneal injection of 5 microg lipopolysaccharide (LPS) were similar in ICE-deficient mice and wild-type (WT) controls. In contrast, plasma ACTH response to restraint or to 200 ng of rat recombinant IL-1beta (rrIL-1beta) was higher in ICE-deficient mice as compared to WT animals. This hyperreactivity of the HPA axis in ICE knockout mice appears not to be related to the production of plasma IL-1beta or IL-6, which was similar to that of WT mice after rrIL-1beta injection. After lipopolysaccharide, ICE-deficient mice exhibited a smaller increase in plasma-immunoreactive IL-1beta and IL-6 as compared to WT controls. After restraint stress neither increase in plasma IL-1beta nor IL-6 was observed. The mechanisms responsible for the increased reactivity of the HPA axis in ICE-deficient mice may result from a higher sensitivity of the HPA axis to inflammatory cytokines or to cleavage products of pro-IL-1beta processed by non-ICE proteases.
Notes:
R M Bluthé, S Layé, B Michaud, C Combe, R Dantzer, P Parnet (2000)  Role of interleukin-1beta and tumour necrosis factor-alpha in lipopolysaccharide-induced sickness behaviour: a study with interleukin-1 type I receptor-deficient mice.   Eur J Neurosci 12: 12. 4447-4456 Dec  
Abstract: Interleukin-1 (IL-1) mediates symptoms of sickness during the host response to infection. IL-1 exerts its effects via several subtypes of receptors. To assess the role of IL-1 receptor type I (IL-1RI) in the sickness-inducing effects of IL-1, IL-1beta and the cytokine inducer lipopolysaccharide were administered to IL-1RI-deficient mice (IL-1RI-/-). Sickness was assessed by depression of social exploration, anorexia, immobility and body weight loss. IL-1RI-/- mice were resistant to the sickness-inducing effects of IL-1beta administered intraperitoneally (2 microg/mouse) and intracerebroventricularly (2 ng/mouse), but still fully responsive to lipopolysaccharide administered intraperitoneally (2.5 microg/mouse) and intracerebroventricularly (3 ng/mouse). The sensitivity of IL-1RI-/- mice to lipopolysaccharide was not due to a higher brain expression of proinflammatory cytokines other than IL-1, since lipopolysaccharide-induced expression of brain IL-1 beta, tumour necrosis factor-alpha (TNF-alpha) and IL-6 transcripts were identical in IL-1RI-/- and control mice when measured by semiquantitative reverse-transcriptase polymerase chain reaction 1 h after treatment. Blockade of TNF-alpha action in the brain by intracerebroventricular administration of a fragment of the soluble TNF receptor, TNF binding protein (3.6 microg/mouse), attenuated the depressive effects of intraperitoneal injection of lipopolysaccharide (1 microg/mouse) on behaviour in IL-1RI-/- but not in control mice. Since IL-1RI-/- mice were not more sensitive to intracerebroventricularly TNF-alpha (50 ng) than control mice, these results indicate that IL-1RI mediates the sickness effect of IL-1 and that TNF-alpha simply replaces IL-1 when this last cytokine is deficient.
Notes:
F Pousset, K Palin, D Verrier, A Bristow, R Dantzer, P Parnet, J Lestage (2000)  Production of interleukin-1 receptor antagonist isoforms by microglia in mixed rat glial cells stimulated by lipopolysaccharide.   Eur Cytokine Netw 11: 4. 682-689 Dec  
Abstract: Although the natural interleukin-1 receptor antagonist (IL-1Ra) has been shown to be produced by microglial cells in response to immune stimuli, nothing was known about the ability of these cells in primary culture to produce the different isoforms of IL-1Ra. Using RT-PCR, we first confirmed that mixed glial cell cultures from newborn rats respond to the cytokine inducer, lipopolysaccharide, by synthesizing IL-1Ra mRNA. Using double immunostaining, we showed that IL-1Ra was detected in microglia but not in astrocytes. Using Western blotting, we finally demonstrated that the IL-1Ra1 isoform was secreted in the supernatant of mixed glial cell cultures, and its production increased in response to lipopolysaccharide. The three different IL-1Ra isoforms were constitutively expressed in cell lysates and their levels increased after lipopolysaccharide treatment, except for IL-1Ra3. These results point to the ability of microglial cells in primary culture to produce the different isoforms of IL-1Ra.
Notes:
1999
F Pousset, S Cremona, R Dantzer, K Kelley, P Parnet (1999)  Interleukin-4 and interleukin-10 regulate IL1-beta induced mouse primary astrocyte activation: a comparative study.   Glia 26: 1. 12-21 Mar  
Abstract: The pro-inflammatory cytokine interleukin-1beta (IL-1beta) is strongly expressed during brain injury and is able to induce severe cellular brain damage via the production of soluble factors. Different processes regulate IL-1 biological activities, like the production of anti-inflammatory cytokines such as interleukin-4 (IL-4) and interleukin-10 (IL-10). In this report, we describe the sequential effects of IL-4 and IL-10 on the production of interleukin-6 (IL-6) induced by IL-1beta in mouse primary astrocytes and compare these effects to those of the synthetic glucocorticoid agonist, dexamethasone. IL-6 secretion and IL-6 mRNA expression were determined by ELISA assay and a comparative RT-PCR method, respectively. Incubation of mouse astrocytes in primary culture simultaneously with IL-1beta (10 ng/ml) + IL-10 (10 ng/ml) or IL-1beta + dexamethasone (10(-6) M) markedly reduced IL-1beta induced IL-6 secretion and IL-6 mRNA expression, respectively, whereas simultaneous addition of IL-4 (10 ng/ml) did not alter the induction of IL-6 by IL-1beta. In contrast, after 24 h of IL-1beta treatment, the level of IL-6 was decreased below constitutive levels, and this change was reversed by addition of IL-4. IL-6 production in IL-1beta pretreated cells was also increased by addition of IL-4, whereas IL-10 and dexamethasone had no effects. The delayed time dependent effect of IL-4 might be partially explained by the induction of IL-4 receptor alpha-chain mRNA expression by IL-1beta. Therefore, we conclude that IL-10 and dexamethasone have rapid immunosuppressive effects on the astrocyte response to IL-1beta stimulation, whereas IL-4, which has a delayed action, acts as an immune inducer.
Notes:
R A French, R W VanHoy, R Chizzonite, J F Zachary, R Dantzer, P Parnet, R M Bluthé, K W Kelley (1999)  Expression and localization of p80 and p68 interleukin-1 receptor proteins in the brain of adult mice.   J Neuroimmunol 93: 1-2. 194-202 Jan  
Abstract: The biological effects of interleukin-1 (IL-1) are mediated by two distinct receptors, the p80 type I IL-1 and p68 type II IL-1 receptor proteins (IL-1RI and IL-1RII, respectively), both of which have been recently co-localized to the growth hormone synthesizing cells of the adenohypophysis. Previous studies have shown that IL-1 can bind to specific structures in the central nervous system, but the distribution of IL-1RI and IL-1RII proteins in the adult mouse brain has not been reported. Here we have used immunohistochemistry to study the expression, distribution and cellular localization of both isoforms of the IL-1 receptor proteins in the adult mouse brain. Using a combination of processing techniques (AMeX fixation and cryosectioning), we have immunolabeled brain sections for each isoform of the IL-1R. Both isoforms are expressed in the CNS, particularly in neuronal soma of the granular layer of the dentate gyrus and pyramidal cells of fields CA1-CA4 of Ammon's horn of the hippocampus, in epithelial cells of the choroid plexus and ependymal layer, and in neuronal soma of Purkinje cells of the cerebellum. The IL-1RII isoform, but not IL-1RI, is expressed in specific neuronal soma and proximal cell processes of neurons of the paraventricular gray matter of the hypothalamus. These immunohistochemical data directly demonstrate the neuronal expression of both IL-1R proteins in situ. The distribution and cellular localization of IL-1R proteins in the CNS provide a molecular basis for understanding reciprocal interactions between the immune system and the brain.
Notes:
1998
S Cremona, S Layé, R Dantzer, P Parnet (1998)  Blockade of brain type II interleukin-1 receptors potentiates IL1beta-induced anorexia in mice.   Neurosci Lett 246: 2. 101-104 Apr  
Abstract: Interleukin-1beta (IL1beta) peripheral activities are mediated by type I IL1 receptors (IL1RI), whereas type II IL1 receptors (IL1RII) act as 'decoy' targets. To study the functionality of IL1RII in the brain, mice were treated with an intracerebroventricular injection of a neutralising MoAb directed against IL1RII (4E2, 1 microg) followed by recombinant rat IL1beta at a dose (2 ng) that produced a moderate but significant decrease of food intake 1 h 30 min after injection. The administration of 4E2 to IL-1beta treated mice significantly potentiated IL1beta-induced decrease in food intake without altering hypothermia. The effects of IL1beta were abrogated in the positive control group treated with IL1ra (2 microg, i.c.v). These results suggest that brain IL1RII down-regulate the effects of IL1beta on its cell targets in the brain.
Notes:
S Cremona, E Goujon, K W Kelley, R Dantzer, P Parnet (1998)  Brain type I but not type II IL-1 receptors mediate the effects of IL-1 beta on behavior in mice.   Am J Physiol 274: 3 Pt 2. R735-R740 Mar  
Abstract: In the immune system, interleukin (IL)-1 beta effects are mediated by the type I IL-1 receptors (IL-1RI), whereas the type II IL-1 receptors (IL-1RII) act as inhibitory receptors. IL-1RI and IL-1RII are also present in the brain. To study their functionality in the brain, mice were centrally treated with neutralizing monoclonal antibody (MAb) directed against IL-1RI (35F5, 1 microgram) or against IL-1RII (4E2, 2 micrograms) and were centrally injected with recombinant rat IL-1 beta at a dose (2 ng) that decreased social exploration. Only 35F5 was effective in abrogating the behavioral effect of IL-1 beta. Moreover, 4E2 (1 microgram i.c.v.) did not potentiate the behavioral response to a subthreshold dose of IL-1 beta (1 ng i.c.v.). To examine the ability of brain IL-1RI to mediate the effects of endogenous IL-1 beta, mice were centrally treated with 35F5 (4 micrograms) and peripherally injected with IL-1 beta (1 microgram). Like IL-1 receptor antagonist (4 micrograms i.c.v.), 35F5 abrogated the effects of IL-1 beta. These results suggest that brain IL-1RI mediates the behavioral effects of IL-1 beta in mice.
Notes:
R Dantzer, R M Bluthé, S Layé, J L Bret-Dibat, P Parnet, K W Kelley (1998)  Cytokines and sickness behavior.   Ann N Y Acad Sci 840: 586-590 May  
Abstract: Peripheral and central injections of interleukin-1 (IL-1) and lipopolysaccharide (LPS) induce the expression of proinflammatory cytokines in the brain and have profound depressing effects on spontaneous and learned behaviors. These effects are mediated by vagal afferents, because they are abrogated by section of the vagus nerves at the subdiaphragmatic level in rats and mice. Vagotomy does not interfere with the synthesis and release of proinflammatory cytokines at the periphery, because plasma and tissue levels of interleukin-1 of vagotomized animals are similar to those of sham-operated animals. Furthermore, the consequences of vagotomy on the host behavioral response to peripheral cytokines are specific to the intraperitoneal route of administration of cytokines because vagotomized animals are still able to respond to IL-1 injected intravenously, subcutaneously, and into the lateral ventricle of the brain. Finally, substance P and cholecystokinin do not appear to play a key role in the transmission of the immune message to the brain because pretreatment by capsaicin or by specific antagonists of CCKA and CCKB receptors does not alter the behavioral effects of LPS and IL-1. All these findings point to the role of neural afferents for transmitting the immune message from the periphery to the brain.
Notes:
R Dantzer, R M Bluthé, G Gheusi, S Cremona, S Layé, P Parnet, K W Kelley (1998)  Molecular basis of sickness behavior.   Ann N Y Acad Sci 856: 132-138 Sep  
Abstract: Peripheral and central injections of lipopolysaccharide (LPS), a cytokine inducer, and recombinant proinflammatory cytokines such as interleukin-1 beta (IL-1 beta) induce sickness behavior in the form of reduced food intake and decreased social activities. Mechanisms of the behavioral effects of cytokines have been the subject of much investigation during the last 3 years. At the behavioral level, the profound depressing effects of cytokines on behavior are the expression of a highly organized motivational state. At the molecular level, sickness behavior is mediated by an inducible brain cytokine compartment that is activated by peripheral cytokines via neural afferent pathways. Centrally produced cytokines act on brain cytokine receptors that are similar to those characterized on peripheral immune and nonimmune cells, as demonstrated by pharmacologic experiments using cytokine receptor antagonists, neutralizing antibodies to specific subtypes of cytokine receptors, and gene targeting techniques. Evidence exists that different components of sickness behavior are mediated by different cytokines and that the relative importance of these cytokines is not the same in the peripheral and central cytokine compartments.
Notes:
1997
T P Bonnert, K E Garka, P Parnet, G Sonoda, J R Testa, J E Sims (1997)  The cloning and characterization of human MyD88: a member of an IL-1 receptor related family.   FEBS Lett 402: 1. 81-84 Jan  
Abstract: Murine MyD88, an RNA with homology both to the interleukin-1 receptor signaling domain and to 'death-domains', is rapidly upregulated during differentiation of the myeloleukemic cell line M1. We have cloned the human homologue of murine MyD88 and re-evaluated the murine sequence. The open reading frame for both species encodes a 296 amino acid protein, which for murine MyD88 is 53 amino acids longer than originally published. Human MyD88 cDNA is encoded by 5 exons, and maps to chromosome 3p21.3-p22 by fluorescence in situ hybridization (FISH). Overexpression of the death domain region leads to transcriptional activation of the IL-8 promoter.
Notes:
E Goujon, S Layé, P Parnet, R Dantzer (1997)  Regulation of cytokine gene expression in the central nervous system by glucocorticoids: mechanisms and functional consequences.   Psychoneuroendocrinology 22 Suppl 1: S75-S80  
Abstract: The proinflammatory cytokines which are released by activated accessory immune cells during the course of an infection have profound effects on the brain. These effects include activation of the hypothalamic-pituitary-adrenal axis, fever and behavioral depression. They are mediated by cytokines which are synthesized and released in the brain, in response to peripherally released cytokines. Glucocorticoids have potent regulatory effects on the synthesis of cytokines by activated macrophages and monocytes. These hormones are also able to regulate the synthesis and action of cytokines in the brain, as demonstrated by the sensitizing effects of adrenalectomy and the depressing effects of stress on the increased cytokine and interleukin-1 beta converting enzyme gene expression that occurs in response to lipopolysaccharide in mice. Preliminary experiments indicate that another way glucocorticoids can contribute to down regulation of the IL-1 system is by increasing the expression of the type II IL-1 receptor in the brain. The regulatory effects of glucocorticoids on cytokine expression in the brain have functional consequences, as demonstrated by the enhanced sensitivity of adrenalectomized animals to the behavioral actions of centrally administered LPS and IL-1. The effects of adrenalectomy are inhibited by compensation with a corticosterone implant and they are mimicked by administration of the type II glucocorticoid receptor, RU 38486. The regulatory role of glucocorticoids on the expression and action of cytokines in the brain makes these hormones and their mechanisms of action key targets for therapeutic interventions in psychopathology and neuropathology.
Notes:
K W Kelley, K Hutchison, R French, R M Bluthé, P Parnet, R W Johnson, R Dantzer (1997)  Central interleukin-1 receptors as mediators of sickness.   Ann N Y Acad Sci 823: 234-246 Aug  
Abstract: These data establish that cytokines, such as IL-1, can act on specific receptors within the brain to induce many symptoms of sickness. A number of inflammatory stimuli in the periphery can activate both the transcription and translation of IL-1 within the central nervous system. It will now be important to determine if similar central IL-1 pathways are activated during SLE and whether these central inflammatory cytokines are involved in the neurologic complications that often accompany this disease.
Notes:
1996
P Parnet, K E Garka, T P Bonnert, S K Dower, J E Sims (1996)  IL-1Rrp is a novel receptor-like molecule similar to the type I interleukin-1 receptor and its homologues T1/ST2 and IL-1R AcP.   J Biol Chem 271: 8. 3967-3970 Feb  
Abstract: A novel member of the interleukin-1 receptor family has been cloned by polymerase chain reaction using degenerate oligonucleotide primers derived from regions of sequence conservation, using as template a yeast artificial chromosome known to contain both interleukin-1 (IL-1) receptors and T1/ST2. The new receptor, called IL-1 receptor-related protein or IL-1Rrp, fails to bind any of the known IL-1 ligands. A chimeric receptor, in which the IL-1Rrp cytoplasmic domain is fused to the extracellular and transmembrane regions of the IL-1 receptor, responds to IL-1 following transfection into COS cells by activation of NFkappaB and induction of IL-8 promoter function.
Notes:
E Goujon, P Parnet, S Layé, C Combe, R Dantzer (1996)  Adrenalectomy enhances pro-inflammatory cytokines gene expression, in the spleen, pituitary and brain of mice in response to lipopolysaccharide.   Brain Res Mol Brain Res 36: 1. 53-62 Feb  
Abstract: To assess the possible influence of endogenous glucocorticoids on cytokine expression in the brain, adrenalectomized mice and sham operated mice were injected with saline or lipopolysaccharide (LPS, 10 micrograms/mouse, subcutaneously) and the levels of transcripts for IL-1 alpha, IL-1 beta, IL-1ra, IL-6 and tumor necrosis factor-alpha (TNF alpha) were determined 2 h after treatment in the spleen, pituitary, hypothalamus, hippocampus and striatum, using semi-quantitative reverse transcription polymerase chain reaction (RT-PCR). Levels of IL-1 beta were measured by ELISA in plasma and tissues of mice sacrificed after the administration of LPS or saline. LPS induced the expression of pro-inflammatory cytokines at the mRNA level in all tissues under investigation, except for TNF alpha in the hippocampus. This effect was potentiated by adrenalectomy in the spleen for IL-1 alpha and IL-1ra, the pituitary for cytokines other than IL-1ra, the hypothalamus for all cytokines, the hippocampus for cytokines other than TNF alpha, and the striatum for IL-1 alpha and IL-6. In saline-treated mice, adrenalectomy increased IL-1 alpha and IL-1 beta gene expression in the hypothalamus and IL-1 alpha gene expression in the hippocampus and striatum. LPS increased plasma and tissue levels of IL-1 beta, as determined by ELISA, and this effect was potentiated by adrenalectomy in plasma and tissues other than the spleen. These results can be interpreted to suggest that endogenous glucocorticoids regulate the neural components of the host response to infection and inflammation by inhibiting cytokine expression in peripheral organs and the brain.
Notes:
J L Mitcham, P Parnet, T P Bonnert, K E Garka, M J Gerhart, J L Slack, M A Gayle, S K Dower, J E Sims (1996)  T1/ST2 signaling establishes it as a member of an expanding interleukin-1 receptor family.   J Biol Chem 271: 10. 5777-5783 Mar  
Abstract: Through data base searches, we have discovered new proteins that share homology with the signaling domain of the type I interleukin-1 receptor (IL-1RI): human "randomly sequenced cDNA 786" (rsc786), murine MyD88, and two partial Drosophila open reading frames, MstProx and STSDm2245. Comparisons between these new proteins and known IL-1RI homologous proteins such as Toll, 18-Wheeler, and T1/ST2 revealed six clusters of amino acid similarity. We tested the hypothesis that sequence similarity between the signaling domain of IL-1RI and the three mammalian family members might indicate functional similarity. Chimeric IL-1RI receptors expressing the putative signaling domains of T1/ST2, MyD88, and rsc786 were assayed by three separate IL-1 responsive assays, NF-kappaB, phosphorylation of an epidermal growth factor receptor peptide, and an interleukin 8 promoter-controlled reporter construct, for their ability to transduce an IL-1-stimulated signal. All three assays were positive in response to the T1/ST2 chimera, while the MyD88 and rsc786 chimeras failed to respond. These data indicate that the sequence homology between IL-1RI and T1/ST2 indicates a functional homology as well.
Notes:
R A French, J F Zachary, R Dantzer, L S Frawley, R Chizzonite, P Parnet, K W Kelley (1996)  Dual expression of p80 type I and p68 type II interleukin-I receptors on anterior pituitary cells synthesizing growth hormone.   Endocrinology 137: 9. 4027-4036 Sep  
Abstract: Interleukin-1 alpha (IL-1 alpha) and IL-1 beta bind to either the p80 type I IL-1 receptor (IL-1RI) or the p68 type II IL-1R (IL-1RII) on both T and B lymphocytes. We and others have previously shown that the anterior pituitary gland also has specific high affinity binding sites for IL-1 alpha (Kd = approximately 1 nM) and expresses transcripts for both isoforms of the IL-1R. However, the identity of cells in the anterior pituitary gland that express the IL-1R and whether different populations of adenohypophyseal cells express different isoforms of the IL-1R remain unknown. Here we have used a combination of immunohistochemistry and histochemistry to localize IL-1RI and IL-1RII to specific target cells in the mouse anterior pituitary gland. Perfusion-fixed, paraffin-embedded sections of anterior pituitary gland were immunolabeled with well characterized monoclonal antibodies to either IL-1RI or IL-1RII and counterstained using a modified Gomori's method to document acidophils and basophils. Immunolabeling demonstrated that both IL-1RI and IL-1RII were abundantly expressed on a single population of anterior pituitary gland cells and that these cells could be classified on the basis of histochemical staining as a subpopulation of acidophils. The distribution, morphology, and proportion of cells immunolabeled for IL-1RI and IL-1RII were consistent with GH-synthesizing cells. To confirm this hypothesis, a modified indirect avidin-biotin complex, sequential peroxidase/alkaline phosphatase technique was used to label anterior pituitary gland cells with antibodies to IL-1RI followed by antibodies to IL-1RII, GH, PRL, or ACTH. The IL-1RI-positive cells predominately coexpressed IL-1RII and GH, but very little, if any, PRL or ACTH. These data establish that the predominant cell population in the murine anterior pituitary gland that constitutively expresses IL-1R stain as acidophils histochemically, is round to oval with dense granular cytoplasm and eccentric nuclei, synthesizes GH, and simultaneously expresses IL-1RI and IL-1RII isoforms.
Notes:
S Layé, E Goujon, C Combe, R VanHoy, K W Kelley, P Parnet, R Dantzer (1996)  Effects of lipopolysaccharide and glucocorticoids on expression of interleukin-1 beta converting enzyme in the pituitary and brain of mice.   J Neuroimmunol 68: 1-2. 61-66 Aug  
Abstract: The present study was carried out to determine whether those factors which regulate the expression of IL-1 beta in immune and non-immune tissues are also able to regulate the expression of ICE. In a first experiment, mice were injected with LPS (10 micrograms/mouse, i.p.) and killed before, 1, 3 or 6 h after the injection. Total RNAs were extracted from the spleen, pituitary and brain (hippocampus and hypothalamus) and submitted to RT-PCR to determine the levels of ICE mRNA as compared to beta 2 microglobulin mRNA. ICE mRNAs were more abundant in the spleen and hippocampus than in the pituitary and hypothalamus but they were not significantly altered by LPS treatment. In a second experiment mice were submitted to adrenalectomy or a 15 min restraint stress and injected with saline or LPS (10 micrograms/mouse. sc). They were killed 1-2 h later and total RNA was extracted from the same tissues as in experiment 1. Adrenalectomized mice had significantly higher ICE mRNA levels whereas stressed mice had significantly lower ICE mRNA levels than their respective controls. These results are discussed with respect to the possible regulatory influence of glucocorticoids on the expression of ICE.
Notes:
1995
E Goujon, P Parnet, A Aubert, G Goodall, R Dantzer (1995)  Corticosterone regulates behavioral effects of lipopolysaccharide and interleukin-1 beta in mice.   Am J Physiol 269: 1 Pt 2. R154-R159 Jul  
Abstract: The modulatory role of endogenous corticoids in the behavioral effects of lipopolysaccharide (LPS) and recombinant human interleukin-1 beta (IL-1 beta) was studied in mice. Adrenalectomy enhanced the depression of social exploration induced by subcutaneous injection of 200 ng of IL-1 beta or 2 micrograms of LPS. This effect was mimicked by an acute injection of the progesterone antagonist RU-38486 (0.25-1 mg). Chronic replacement with a 15-mg corticosterone pellet abrogated the enhanced susceptibility of adrenalectomized animals to 200 ng of IL-1 beta but had only partial protective effects on their response to 400 ng of IL-1 beta and LPS. These results suggest that the pituitary-adrenal response to cytokines exerts an inhibitory feedback on the cell targets that mediate the behavioral effects of LPS and IL-1 beta.
Notes:
S Layé, R M Bluthé, S Kent, C Combe, C Médina, P Parnet, K Kelley, R Dantzer (1995)  Subdiaphragmatic vagotomy blocks induction of IL-1 beta mRNA in mice brain in response to peripheral LPS.   Am J Physiol 268: 5 Pt 2. R1327-R1331 May  
Abstract: To test the possibility that the vagus nerve is involved in the communication between the immune system and the brain, we injected sham-operated and vagotomized mice with physiological saline or lipopolysaccharide (LPS; 400 micrograms/kg ip). Vagotomy attenuated LPS-induced depression of general activity measured 2 h after treatment but did not alter the increase in plasma levels of IL-1 beta in response to LPS. In addition, vagotomy abrogated the LPS-induced increase in the levels of transcripts for IL-1 beta, as determined by semiquantitative polymerase chain reaction after reverse transcription, in the hypothalamus and hippocampus, but not in the pituitary of vagotomized mice. This relationship between the effects of vagotomy on the behavioral effects of LPS and the LPS-induced brain expression of IL-1 beta mRNA indicates that vagal afferent fibers play a prominent role in the pathways of communication between the immune system and the brain.
Notes:
E Goujon, P Parnet, S Laye, C Combe, K W Kelley, R Dantzer (1995)  Stress downregulates lipopolysaccharide-induced expression of proinflammatory cytokines in the spleen, pituitary, and brain of mice.   Brain Behav Immun 9: 4. 292-303 Dec  
Abstract: Mice injected with LPS (10 mu g/mouse, sc) or saline were submitted to a 15-min restraint stress and sacrificed 1 or 2 h later to assess the effect of stress on the induction of interleukin-1beta (IL-1beta) and other proinflammatory cytokines (IL-1alpha, IL-1ra, IL-6, and tumor necrosis factor-alpha) in the spleen, pituitary, hypothalamus, hippocampus, and striatum. LPS-induced cytokine gene expression, as determined by comparative RT-PCR, was lower in stressed than in nonstressed mice. LPS increased plasma and tissue levels of IL-1beta, as determined by ELISA, but this effect was less marked in stressed than in nonstressed mice. These results are discussed in relation to the modulatory effects of glucocorticoids on cytokine production.
Notes:
E Goujon, P Parnet, S Cremona, R Dantzer (1995)  Endogenous glucocorticoids down regulate central effects of interleukin-1 beta on body temperature and behaviour in mice.   Brain Res 702: 1-2. 173-180 Dec  
Abstract: Adrenalectomy sensitizes laboratory animals to the pyrogenic and behavioural effects of proinflammatory cytokines. To determine whether these effects are mediated by central sites of action of glucocorticoids, interleukin-1 beta was injected intracerebroventricularly (i.c.v.) in adrenalectomized mice with or without corticosterone supplementation and in mice pretreated i.c.v. with the glucocorticoid type II receptor antagonist RU38486. Adrenalectomized mice were more sensitive to the depressing effects of i.c.v. IL-1 beta on body temperature and social exploration than sham-operated mice. Corticosterone supplementation reversed the increased sensitivity to the low (300 pg/mouse) but not to the high dose (900 pg/mouse) of IL-1 beta. Central administration of RU38486 (0.5-1 microgram/mouse) mimicked the effects of adrenalectomy on behaviour but not on body temperature. These results suggest that endogenous glucocorticoids released in response to IL-1 beta act in the brain to modulate the sensitivity of the cellular targets of this cytokine.
Notes:
1994
Q Liu, S Arkins, A Biragyn, C Minshall, P Parnet, R Dantzer, K W Kelley (1994)  Competitive reverse transcriptase-polymerase chain reaction using a synthetic internal RNA standard to quantitate transcripts for leukocyte-derived hormones.   Neuroimmunomodulation 1: 1. 33-41 Jan  
Abstract: Leukocytes synthesize a variety of hormones that were once thought to be unique products of endocrine tissues. Understanding the regulation of leukocyte-derived hormone synthesis requires an accurate means for measuring steady-state expression of specific mRNA transcripts. Here we describe a competitive reverse transcriptase-polymerase chain reaction (RT-PCR) technique to accurately quantitate macrophage-derived insulin-like growth factor-I (IGF-I) mRNA, and demonstrate the utility of this approach for measuring expression of leukocyte-derived hormone transcripts. A riboprobe was constructed to generate approximately 1 kb of synthetic competitor IGF-I RNA (exons 1 and 3-6) that differed from cellular IGF-I RNA by insertion of 122 bp of beta-actin RNA. One set of oligonucleotide primers could thus be used to simultaneously reverse transcribe and amplify both 144 bp of cellular (exons 3 and 4) and 266 bp of competitor IGF-I RNA. Densitometric scanning of the PAGE-separated PCR products revealed that the ratio of competitor to cellular amplified DNA bore a linear relationship (r2 > or = 0.98) to the amount of competitor RNA for both rat liver and splenocytes. However, rat liver contained 104 x 10(6) IGF-I molecules per microgram of total cellular RNA compared to only 2 x 10(6) IGF-I molecules for splenocytes.(ABSTRACT TRUNCATED AT 250 WORDS)
Notes:
R M Bluthé, V Walter, P Parnet, S Layé, J Lestage, D Verrier, S Poole, B E Stenning, K W Kelley, R Dantzer (1994)  Lipopolysaccharide induces sickness behaviour in rats by a vagal mediated mechanism.   C R Acad Sci III 317: 6. 499-503 Jun  
Abstract: To assess the possibility that lipopolysaccharide (LPS) induces sickness behaviour by activating primary afferent nerves, the effects of LPS (1.25 mg kg-1, intraperitoneally) were compared in vagotomized and sham-operated rats. Subdiaphragmatic vagotomy blocked the LPS-induced depression of social investigation but had no effect on LPS-induced increases in levels of IL-1 beta in plasma and peritoneal macrophages and on LPS-induced changes in dehydrogenase activity of peritoneal macrophages.
Notes:
R M Bluthé, M Pawlowski, S Suarez, P Parnet, Q Pittman, K W Kelley, R Dantzer (1994)  Synergy between tumor necrosis factor alpha and interleukin-1 in the induction of sickness behavior in mice.   Psychoneuroendocrinology 19: 2. 197-207  
Abstract: Like interleukin-1, recombinant human tumor necrosis factor alpha (TNF alpha) has been found to decrease social exploration and induce weight loss in mice in a dose and time-dependent manner. The present study was carried out to study the interaction between these two cytokines. Mice were injected IP with subthreshold doses of TNF alpha (2.5 micrograms/mouse) and IL-1 beta (50 ng/mouse). Social exploration was decreased 2 and 4 h after injection of TNF and IL-1, but body weight was not affected. Subthreshold doses of TNF alpha (90 ng/mouse) and IL-1 beta (100 pg/mouse) were also injected intracerebroventricularly (ICV). Social exploration was decreased 1.5 and 3 h after injections of the two cytokines and body weight was decreased for 6 h. To test the possibility of central induction of IL-1 by TNF alpha, mice pretreated with IL-1 receptor antagonist (IL-1ra, 1.8 micrograms/mouse, ICV) were injected with 90 ng TNF alpha. Pretreatment with IL-1ra antagonized the depressive effect of TNF alpha on behavior, but had no effect on weight loss induced by this cytokine. These results suggest that TNF alpha-induced behavioral alterations are mediated by endogenously released IL-1, whereas metabolic changes are dependent on the release of other cytokines.
Notes:
S Layé, P Parnet, E Goujon, R Dantzer (1994)  Peripheral administration of lipopolysaccharide induces the expression of cytokine transcripts in the brain and pituitary of mice.   Brain Res Mol Brain Res 27: 1. 157-162 Nov  
Abstract: The reverse transcription polymerase chain reaction (RT-PCR) was used to assess the induction of mRNA of the proinflammatory cytokines IL-1 beta, IL-6 and TNF alpha in the spleen, pituitary, hypothalamus and hippocampus of mice after an intraperitoneal injection of lipopolysaccharide (LPS, 10 micrograms/mouse). The kinetics of cytokine gene expression induced by peripheral LPS in the pituitary and brain structures were different from that observed in the spleen. For IL-1 beta the dose-response curve was also measured and also found to be different. These results support the idea that one pathway by which peripheral immune stimuli affect brain functions includes local synthesis of proinflammatory cytokines in certain brain structures.
Notes:
P Parnet, S Amindari, C Wu, D Brunke-Reese, E Goujon, J A Weyhenmeyer, R Dantzer, K W Kelley (1994)  Expression of type I and type II interleukin-1 receptors in mouse brain.   Brain Res Mol Brain Res 27: 1. 63-70 Nov  
Abstract: Although binding sites for IL-1 have been identified in the mouse brain, it is still unknown whether these binding sites correspond to the type I or type II IL-1 receptor. Quantitative autoradiography was used to confirm the presence of specific binding sites for radiolabelled recombinant human IL-1 alpha (125I-HuIL-1 alpha) in the brain of DBA/2 mice. IL-1 binding was highest in the dentate gyrus, consisting of a single class of high affinity binding sites with a Kd of 0.1 nM and a Bmax of 57 fmol/mg protein. A similar Kd of 0.2 nM was obtained using isolated membranes from the whole hippocampus, although the number of binding sites was lower (2 fmol/mg protein). Affinity cross-linking of 125I-Hu-IL-1 alpha to hippocampal membranes revealed the existence of two types of IL-1 receptor proteins, consistent with the sizes of the type I (85 kD) and type II (60 kD) IL-1 receptor. Oligonucleotide probes were then synthesized and used in RT-PCR followed by Southern blotting to show that the whole brain expresses transcripts for both the type I and type II IL-1 receptors. The murine neuroblastoma cell line, C1300, expresses type I rather than type II IL-1 receptor mRNA. The type I receptor protein can be identified by flow cytometry on the membrane of the C1300 neuronal cell line using indirect immunofluorescence with a rat anti-mouse type I IL-1 receptor MoAb. These data show that mouse brain expresses both type I and type II IL-1 receptor mRNA and proteins and offer further support to the idea that type I IL-1 receptors are synthesized and expressed by neurons.
Notes:
1993
P Parnet, D L Brunke, E Goujon, J D Mainard, A Biragyn, S Arkins, R Dantzer, K W Kelley (1993)  Molecular identification of two types of interleukin-1 receptors in the murine pituitary gland.   J Neuroendocrinol 5: 2. 213-219 Apr  
Abstract: The present study was carried out to characterize interleukin-1 (IL-1) receptors on murine pituitary cells. Receptor autoradiography confirmed the existence of binding sites for IL-1 alpha in the murine adenohypophysis, but not in the neural or intermediate lobes. Specific binding of IL-1 to isolated pituitary membranes revealed a Kd of 0.9 nM with a Bmax of 37 fmol/mg protein. To examine the possibility that the adenohypophysis synthesizes a receptor for IL-1, immunocytochemistry experiments with a specific monoclonal antibody against the type I receptor revealed the existence of this protein in only the adenohypophysis. Identity of the type I IL-1 receptor was similar to that found on T cells as determined by: 1) amplification of the predicted 619 bp fragment spanning the cytoplasmic, transmembrane and extracellular domains from RNA of pituitary and T cell origin, as well as clonal AtT-20 pituitary cells, and 2) restriction fragment analysis and sequencing of the amplified cDNAs. The pituitary gland and AtT-20 cells also expressed transcripts for the newly identified type II receptor for IL-1 as assessed by amplification of a specific 325 bp fragment, restriction fragment analysis and nucleotide sequencing, and these transcripts were similar to those found on B lymphocytes. These data identify two different forms of the IL-1 receptor in both normal and transformed pituitary cells and establish that these receptors are similar at the molecular level to those first identified on T and B lymphocytes.
Notes:
1991
P Parnet, M Mitsuhashi, C W Turck, B Kerdelhue, D G Payan (1991)  Tachykinin receptor cross-talk. Immunological cross-reactivity between the external domains of the substance K and substance P receptors.   Brain Behav Immun 5: 1. 73-83 Mar  
Abstract: In the present study, we have chemically synthesized peptides which correspond to the four putative extracellular domains of the predicted substance K (SK) receptor protein and raised specific polyclonal antibodies against these peptides. These antibodies were then tested for both structural and functional recognition of epitopes on the substance P (SP) receptor on rat AR42J pancreatic cells and human IM9 lymphoblasts, which express the SP receptor, but not the SK receptor. Antibodies directed against the first, second, and fourth external domains of the predicted SK receptor recognized a 58-kDa protein on AR42J cells. This protein has a molecular weight similar to the previously demonstrated SP receptor on both AR42J cells and IM9 cells. Furthermore, antibodies against the second and fourth extracellular domains significantly inhibited specific 125I-SP binding on both AR42J and IM9 cells, and also significantly inhibited SP-induced mobilization of [Ca2+]i on AR42J cells. These data suggest that the second and fourth extracellular domains of the SK and SP receptors may share common structural motifs for ligand binding and signaling mechanism.
Notes:
1990
P Parnet, D G Payan, B Kerdelhué, M Mitsuhashi (1990)  Neuro-endocrine interaction on lymphocytes. Testosterone-induced modulation of the lymphocyte substance P receptor.   J Neuroimmunol 28: 2. 185-188 Jul  
Abstract: Human IM-9 B-lymphoblasts have been shown to express the receptor for the neuropeptide substance P (SP). The present study was undertaken to evaluate the effect of sex hormones on this receptor. Testosterone inhibited [125I]SP binding in a dose-dependent manner with an IC50 of approximately 100 nM, while both estradiol and progesterone failed to inhibit SP binding even at concentrations as high as 1 microM. Furthermore, Scatchard analysis indicated that the dissociation constant (Kd) of the SP receptor was markedly increased from 0.25 nM to 2.2 nM when cells were incubated with testosterone. These data indicate a unique neuro-endocrine interaction on lymphocytes involving the substance P receptor.
Notes:
P Parnet, V Lenoir, M Palkovits, B Kerdelhué (1990)  Estrous cycle variations in gonadotropin-releasing hormone, substance p and Beta-endorphin contents in the median eminence, the arcuate nucleus and the medial preoptic nucleus in the rat: a detailed analysis of proestrus changes.   J Neuroendocrinol 2: 3. 291-296 Jun  
Abstract: Abstract The concentrations of gonadotropin-releasing hormone, substance P and beta-endorphin were measured in the median eminence, the arcuate nucleus and the medial preoptic nucleus of 4-day cycling female rats. Very well marked estrous cycle-related fluctuations were registered for these neuropeptides in these areas. The largest variations in concentrations of peptide levels were observed in the median eminence. Substance P concentration was highest throughout the day of proestrus as compared to the three others days of the cycle. At the time of the preovulatory luteinizing hormone surge, on the afternoon of proestrus, there was a marked increase in gonadotropin-releasing hormone concentration and a marked decrease in beta-endorphin concentration.
Notes:
1988
B Kerdelhué, P Parnet, V Lenoir, A Schirar, F Gaudoux, M C Levasseur, M Palkovits, C Blacker, R Scholler (1988)  Interactions between 17 beta-estradiol and the hypothalamo-pituitary beta-endorphin system in the regulation of the cyclic LH secretion.   J Steroid Biochem 30: 1-6. 161-168  
Abstract: This paper further substantiates the physiological role of beta-endorphin (beta-END) in the control of the cyclic LH secretion and provides new data on the interactions between 17 beta-estradiol (17 beta-E2) and beta-END at both the hypothalamic and pituitary levels. At the hypothalamic level, during the estrous cycle in rats, beta-END concentrations were highest on diestrus I in the arcuate nucleus, median preoptic area and median eminence and lowest at the time of the preovulatory 17 beta-E2 surge on proestrus, before the subsequent preovulatory hypothalamic GnRH and plasma LH surges. Data obtained in ovariectomized 17 beta-E2-treated ewes support the direct involvement of 17 beta-E2 in changes in beta-END and GnRH concentrations in these hypothalamic areas. At the anterior pituitary level, in vitro results obtained using anterior pituitaries from the proestrus morning cycling female rat have shown that 17 beta-E2 strongly suppresses beta-END secretion and that GnRH stimulates the release of beta-END. Furthermore, marked fluctuations were observed for plasma beta-END throughout the menstrual cycle in the woman. Low beta-END concentrations were observed in the period preceding the LH preovulatory surge. Taken together, these results show that: (1) decreases in hypothalamic beta-END concentrations, which are controlled at least by circulating levels of 17 beta-E2, modulate GnRH synthesis and/or release and contribute to the mechanisms which initiate the LH surge; (2) anterior pituitary beta-END might be involved in the mechanisms which terminate the LH surge.
Notes:
Powered by PublicationsList.org.