Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Eur J Immunol ; 46(10): 2467-2480, 2016 10.
Article in English | MEDLINE | ID: mdl-27457277

ABSTRACT

Tertiary lymphoid tissue (TLT) is lymphoid tissue that forms in adult life as a result of chronic inflammation in a tissue or organ. TLT has been shown to form in a variety of chronic inflammatory diseases, though it is not clear if and how TLT develops in the inflamed colon during inflammatory bowel disease. Here, we show that TLT develops as newly formed lymphoid tissue in the colon following dextran sulphate sodium induced colitis in C57BL/6 mice, where it can be distinguished from the preexisting colonic patches and solitary intestinal lymphoid tissue. TLT in the inflamed colon develops following the expression of lymphoid tissue-inducing chemokines and adhesion molecules, such as CXCL13 and VCAM-1, respectively, which are produced by stromal organizer cells. Surprisingly, this process of TLT formation was independent of the lymphotoxin signaling pathway, but rather under neuronal control, as we demonstrate that selective surgical ablation of vagus nerve innervation inhibits CXCL13 expression and abrogates TLT formation without affecting colitis. Sympathetic neuron denervation does not affect TLT formation. Hence, we reveal that inflammation in the colon induces the formation of TLT, which is controlled by innervation through the vagus nerve.


Subject(s)
Colitis/immunology , Colon/innervation , Lymphoid Tissue/innervation , Tertiary Lymphoid Structures/pathology , Vagus Nerve/pathology , Animals , Chemokine CXCL13/genetics , Chemokine CXCL13/metabolism , Colitis/chemically induced , Colon/pathology , Dextran Sulfate , Female , Lymphoid Tissue/pathology , Lymphotoxin-alpha/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Vascular Cell Adhesion Molecule-1/metabolism
2.
Am J Physiol Heart Circ Physiol ; 309(4): H646-54, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26092978

ABSTRACT

UNLABELLED: The brain plays a prominent role in the regulation of inflammation. Immune cells are under control of the so-called cholinergic anti-inflammatory reflex, mainly acting via autonomic innervation of the spleen. Activation of this reflex inhibits the secretion of proinflammatory cytokines and may reduce the development of atherosclerosis. Therefore, the aim of this study was to evaluate the effects of selective parasympathetic (Px) and sympathetic (Sx) denervation of the spleen on inflammatory status and atherosclerotic lesion development. Female APOE*3-Leiden.CETP mice, a well-established model for human-like lipid metabolism and atherosclerosis, were fed a cholesterol-containing Western-type diet for 4 wk after which they were subdivided into three groups receiving either splenic Px, splenic Sx, or sham surgery. The mice were subsequently challenged with the same diet for an additional 15 wk. Selective Px increased leukocyte counts (i.e., dendritic cells, B cells, and T cells) in the spleen and increased gene expression of proinflammatory cytokines in the liver and peritoneal leukocytes compared with Sx and sham surgery. Both Px and Sx increased circulating proinflammatory cytokines IL-1ß and IL-6. However, the increased proinflammatory status in denervated mice did not affect atherosclerotic lesion size or lesion composition. CONCLUSION: Predominantly selective Px of the spleen enhances the inflammatory status, which, however, does not aggravate diet-induced atherosclerotic lesion development.


Subject(s)
Atherosclerosis/physiopathology , Autonomic Nervous System/physiology , Spleen/immunology , Animals , Apolipoprotein E3/genetics , Atherosclerosis/etiology , Atherosclerosis/immunology , Denervation , Diet, High-Fat/adverse effects , Female , Inflammation/immunology , Inflammation/physiopathology , Interleukin-1beta/blood , Interleukin-6/blood , Mice , Reflex , Spleen/innervation
3.
Gut ; 63(6): 938-48, 2014 Jun.
Article in English | MEDLINE | ID: mdl-23929694

ABSTRACT

The cholinergic anti-inflammatory pathway (CAIP) has been proposed as a key mechanism by which the brain, through the vagus nerve, modulates the immune system in the spleen. Vagus nerve stimulation (VNS) reduces intestinal inflammation and improves postoperative ileus. We investigated the neural pathway involved and the cells mediating the anti-inflammatory effect of VNS in the gut. The effect of VNS on intestinal inflammation and transit was investigated in wild-type, splenic denervated and Rag-1 knockout mice. To define the possible role of α7 nicotinic acetylcholine receptor (α7nAChR), we used knockout and bone marrow chimaera mice. Anterograde tracing of vagal efferents, cell sorting and Ca(2+) imaging were used to reveal the intestinal cells targeted by the vagus nerve. VNS attenuates surgery-induced intestinal inflammation and improves postoperative intestinal transit in wild-type, splenic denervated and T-cell-deficient mice. In contrast, VNS is ineffective in α7nAChR knockout mice and α7nAChR-deficient bone marrow chimaera mice. Anterograde labelling fails to detect vagal efferents contacting resident macrophages, but shows close contacts between cholinergic myenteric neurons and resident macrophages expressing α7nAChR. Finally, α7nAChR activation modulates ATP-induced Ca(2+) response in small intestine resident macrophages. We show that the anti-inflammatory effect of the VNS in the intestine is independent of the spleen and T cells. Instead, the vagus nerve interacts with cholinergic myenteric neurons in close contact with the muscularis macrophages. Our data suggest that intestinal muscularis resident macrophages expressing α7nAChR are most likely the ultimate target of the gastrointestinal CAIP.


Subject(s)
Macrophages/metabolism , Muscle, Smooth/cytology , Vagus Nerve Stimulation , Vagus Nerve/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Autonomic Denervation , Cytokines/genetics , Enteritis/metabolism , Gastrointestinal Transit , Gene Expression , Macrophages/cytology , Mice , Mice, Knockout , Myenteric Plexus/metabolism , Neurons/metabolism , Nicotine/pharmacology , Peroxidase/metabolism , Signal Transduction , Spleen/innervation , alpha7 Nicotinic Acetylcholine Receptor/agonists , alpha7 Nicotinic Acetylcholine Receptor/genetics
4.
Gut ; 62(11): 1581-90, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23242119

ABSTRACT

OBJECTIVE: Intestinal inflammation resulting from manipulation-induced mast cell activation is a crucial mechanism in the pathophysiology of postoperative ileus (POI). Recently it has been shown that spleen tyrosine kinase (Syk) is involved in mast cell degranulation. Therefore, we have evaluated the effect of the Syk-inhibitor GSK compound 143 (GSK143) as potential treatment to shorten POI. DESIGN: In vivo: in a mouse model of POI, the effect of the Syk inhibitor (GSK143) was evaluated on gastrointestinal transit, muscular inflammation and cytokine production. In vitro: the effect of GSK143 and doxantrazole were evaluated on cultured peritoneal mast cells (PMCs) and bone marrow derived macrophages. RESULTS: In vivo: intestinal manipulation resulted in a delay in gastrointestinal transit at t=24 h (Geometric Center (GC): 4.4 ± 0.3). Doxantrazole and GSK143 significantly increased gastrointestinal transit (GC doxantrazole (10 mg/kg): 7.2 ± 0.7; GSK143 (1 mg/kg): 7.6 ± 0.6), reduced inflammation and prevented recruitment of immune cells in the intestinal muscularis. In vitro: in PMCs, substance P (0-90 µM) and trinitrophenyl (0-4 µg/ml) induced a concentration-dependent release of ß-hexosaminidase. Pretreatment with doxantrazole and GSK143 (0.03-10 µM) concentration dependently blocked substance P and trinitrophenyl induced ß-hexosaminidase release. In addition, GSK143 was able to reduce cytokine expression in endotoxin-treated bone marrow derived macrophages in a concentration-dependent manner. CONCLUSIONS: The Syk inhibitor GSK143 reduces macrophage activation and mast cell degranulation in vitro. In addition, it inhibits manipulation-induced intestinal muscular inflammation and restores intestinal transit in mice. These findings suggest that Syk inhibition may be a new tool to shorten POI.


Subject(s)
Aniline Compounds/therapeutic use , Ileus/prevention & control , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Postoperative Complications/prevention & control , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/therapeutic use , Aniline Compounds/administration & dosage , Aniline Compounds/pharmacology , Animals , Cell Degranulation/drug effects , Cells, Cultured , Cytokines/biosynthesis , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Gastrointestinal Transit/drug effects , Ileus/physiopathology , Macrophage Activation/drug effects , Mast Cells/drug effects , Mast Cells/physiology , Mice , Mice, Inbred C57BL , Ovalbumin/antagonists & inhibitors , Ovalbumin/pharmacology , Phosphodiesterase Inhibitors/therapeutic use , Postoperative Complications/physiopathology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Substance P/antagonists & inhibitors , Substance P/pharmacology , Syk Kinase , Thioxanthenes/therapeutic use , Xanthones/therapeutic use
5.
Am J Physiol Gastrointest Liver Physiol ; 305(5): G383-91, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23812038

ABSTRACT

Nutritional stimulation of the cholecystokinin-1 receptor (CCK-1R) and nicotinic acetylcholine receptor (nAChR)-mediated vagal reflex was shown to reduce inflammation and preserve intestinal integrity. Mast cells are important early effectors of the innate immune response; therefore modulation of mucosal mast cells is a potential therapeutic target to control the acute inflammatory response in the intestine. The present study investigates intestinal mast cell responsiveness upon nutritional activation of the vagal anti-inflammatory reflex during acute inflammation. Mucosal mast cell degranulation was induced in C57/Bl6 mice by administration of Salmonella enterica LPS. Lipid-rich enteral feeding prior to LPS significantly decreased circulatory levels of mouse mast cell protease at 30 min post-LPS compared with isocaloric low-lipid nutrition or fasting. CCK-1R blockage reversed the inhibitory effects of lipid-rich feeding, whereas stimulation of the peripheral CCK-1R mimicked nutritional mast cell inhibition. The effects of lipid-rich nutrition were negated by nAChR blockers chlorisondamine and α-bungarotoxin and vagal intestinal denervation. Accordingly, release of ß-hexosaminidase by MC/9 mast cells following LPS or IgE-ovalbumin complexes was dose dependently inhibited by acetylcholine and nicotine. Application of GSK1345038A, a specific agonist of the nAChR α7, in bone marrow-derived mast cells from nAChR ß2-/- and wild types indicated that cholinergic inhibition of mast cells is mediated by the nAChR α7 and is independent of the nAChR ß2. Together, the present study reveals mucosal mast cells as a previously unknown target of the nutritional anti-inflammatory vagal reflex.


Subject(s)
Cell Degranulation , Dietary Fats/administration & dosage , Enteral Nutrition , Inflammation/prevention & control , Intestinal Mucosa/immunology , Intestinal Mucosa/innervation , Mast Cells/immunology , Reflex , Vagus Nerve/physiopathology , Animals , Cell Degranulation/drug effects , Cell Line , Cholinergic Agonists/pharmacology , Chymases/blood , Disease Models, Animal , Histamine Antagonists/pharmacology , Immunity, Mucosal , Inflammation/blood , Inflammation/immunology , Inflammation/physiopathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Lipopolysaccharides , Male , Mast Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Nicotinic Antagonists/pharmacology , Receptor, Cholecystokinin A/metabolism , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Vagotomy, Proximal Gastric , Vagus Nerve/drug effects , Vagus Nerve/immunology , Vagus Nerve/metabolism , Vagus Nerve/surgery , beta-N-Acetylhexosaminidases/metabolism
6.
Eur J Neurosci ; 30(9): 1665-75, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19878276

ABSTRACT

Food-anticipatory activity (FAA) and especially the food-entrained oscillator (FEO) have driven many scientists to seek their mechanisms and locations. Starting our research on FAA we, possibly like many other scientists, were convinced that clock genes held the key to the location and the underlying mechanisms for FAA. In this review, which is aimed especially at discussing the contribution of the peripheral oscillators, we have put together the accumulating evidence that the clock gene machinery as we know it today is not sufficient to explain food entrainment. We discuss the contribution of three types of oscillating processes: (i) within the suprachiasmatic nucleus (SCN), neurons capable of maintaining a 24-h oscillation in electrical activity driven by a set of clock genes; (ii) oscillations in metabolic genes and clock genes in other parts of the brain and in peripheral organs driven by the SCN or by food, which damp out after a few cycles; (iii) an FEO which, we propose, is a system built up of different oscillatory processes and consisting of an as-yet-unidentified network of central and peripheral structures. In view of the evidence that clock genes and metabolic oscillations are not essential for the persistence of FAA we propose that food entrainment is initiated by a repeated metabolic state of scarcity that drives an oscillating network of brain nuclei in interaction with peripheral oscillators. This complex may constitute the proposed FEO and is distributed in our peripheral organs as well as within the central nervous system.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , Feeding Behavior/physiology , Animals , Behavior, Animal/physiology , Biological Clocks/genetics , Circadian Rhythm/genetics , Eating/physiology , Food , Germinal Center Kinases , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Light , Neurons/metabolism , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Suprachiasmatic Nucleus/cytology , Suprachiasmatic Nucleus/physiology , Time Perception/physiology
7.
Endocrinology ; 149(4): 1914-25, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18162516

ABSTRACT

Daily variations in plasma glucose concentrations are controlled by the biological clock, located in the suprachiasmatic nucleus. Our previous studies indicated an important role for the sympathetic innervation of the liver in the generation of the daily glucose rhythm. In the present study, we investigated further the role of the autonomic nervous system (ANS) in the genesis of the plasma glucose rhythm. First, we showed that complete removal of the autonomic inputs to the liver did not impair the plasma glucose rhythm or the daily expression of the glucoregulatory enzymes in the liver. Consequently, we studied whether the daily glucose rhythm is driven by the daily feeding activity in denervated animals. Surprisingly, complete denervation combined with a noncircadian feeding schedule also did not abolish the 24-h profile in plasma glucose or all daily rhythms in the gene expression of liver enzymes. These results demonstrate that the mechanisms used by the suprachiasmatic nucleus to control the rhythmic expression of glucose-metabolizing enzymes and the 24-h rhythm in plasma glucose concentrations are highly versatile and the glucose rhythm can be maintained in absence of hepatic ANS input and/or a day/night rhythm in feeding activity. Interestingly, a hepatic sympathectomy or parasympathectomy did abolish the plasma glucose rhythm, demonstrating that a unilateral denervation of the liver is more deleterious to maintaining the rhythmic liver metabolism than a complete removal of both branches. This observation supports the notion that an unbalanced ANS in obesity and diabetes accounts for the disturbed glucose balance in these disorders.


Subject(s)
Autonomic Nervous System/physiology , Blood Glucose/analysis , Circadian Rhythm/physiology , Liver/enzymology , Liver/innervation , Animals , Corticosterone/blood , Glucose/metabolism , Insulin/blood , Liver Glycogen/analysis , Male , RNA, Messenger/analysis , Rats , Rats, Wistar , Suprachiasmatic Nucleus/physiology
8.
F1000Res ; 5: 98, 2016.
Article in English | MEDLINE | ID: mdl-26925229

ABSTRACT

BACKGROUND: Enhanced colorectal sensitivity (i.e. visceral hypersensitivity) is thought to be a pathophysiological mechanism in irritable bowel syndrome (IBS). In healthy men a circadian variation in rectal perception to colonic distention was described. Disturbed day and night rhythms, which occur in shift work and trans meridian flights, are associated with the prevalence of IBS. This raises the question whether disruptions of circadian control are responsible for the observed pathology in IBS. Prior to investigating altered rhythmicity in relation to visceral hypersensitivity in a rat model for IBS, it is relevant to establish whether normal rats display circadian variation similar to healthy men.  METHODOLOGY AND FINDINGS: In rodents colorectal distension leads to reproducible contractions of abdominal musculature. We used quantification of this so called visceromotor response (VMR) by electromyography (EMG) to assess visceral sensitivity in rats. We assessed the VMR in normal male Long Evans rats at different time points of the light/dark cycle. Although a control experiment with male maternal separated rats confirmed that intentionally inflicted (i.e. stress induced) changes in VMR can be detected, normal male Long Evans rats showed no variation in VMR along the light/dark cycle in response to colorectal distension. CONCLUSIONS: In the absence of a daily rhythm of colorectal sensitivity in normal control rats it is not possible to investigate possible aberrancies in our rat model for IBS.

9.
Mol Cell Endocrinol ; 422: 125-131, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26704081

ABSTRACT

The master clock in the hypothalamic suprachiasmatic nucleus (SCN) is assumed to distribute rhythmic information to the periphery via neural, humoral and/or behavioral connections. Until now, feeding, corticosterone and neural inputs are considered important signals for synchronizing daily rhythms in the liver. In this study, we investigated the necessity of neural inputs as well as of the feeding and adrenal hormone rhythms for maintaining daily hepatic clock gene rhythms. Clock genes kept their daily rhythm when only one of these three signals was disrupted, or when we disrupted hepatic neuronal inputs together with the adrenal hormone rhythm or with the daily feeding rhythm. However, all clock genes studied lost their daily expression rhythm after simultaneous disruption of the feeding and adrenal hormone rhythm. These data indicate that either a daily rhythm of feeding or adrenal hormones should be present to synchronize clock gene rhythms in the liver with the SCN.


Subject(s)
CLOCK Proteins/metabolism , Corticosterone/metabolism , Feeding Behavior/physiology , Liver/metabolism , Neurons/metabolism , Animals , Circadian Clocks , Gene Expression Profiling , Gene Expression Regulation , Male , Rats , Rats, Wistar , Suprachiasmatic Nucleus/metabolism
10.
Neuro Endocrinol Lett ; 24(1-2): 33-8, 2003.
Article in English | MEDLINE | ID: mdl-12743529

ABSTRACT

OBJECTIVES: The pineal gland transduces photoperiodic informations to the neuroendocrine axis through the nocturnally melatonin secretion. This hormonal message plays a major role in the biological rhythm regulation. By autoradiography, more than 130 melatonin putative targets have been reported in the central nervous system (CNS) and in peripheral tissues. However, cross-species consensus concern only a few of them like the suprachiasmatic nuclei (SCN), the master circadian clock, and the pars tuberalis of the pituitary. Recently, MT1 melatonin receptor cDNA have been cloned in several mammals providing us with new tools to investigate its tissular location at the gene level. In the present study, we report a screening for MT1 mRNA by RT-PCR amplification of numerous tissue mRNA. METHOD: mRNA were extracted from a large variety of rat tissues. To semi-quantify the melatonin receptor mRNA expression level, each cDNA was amplified concomitantly with both beta-actin and MT1 specific primers. RESULTS: In central and peripheral tissues previously reported to bind melatonin, strong PCR signals were logically observed. More surprisingly, a vast majority of studied tissues express MT1 mRNA and then might be responsive to melatonin. CONCLUSION: Numerous biological functions express diurnal rhythmicity and internal-synchronization. As, most of them apparently do not receive any out-coming neuronal message from the SCN, endocrine communication was proposed to support biological rhythm synchronization. Our present data strengthen the idea that the nocturnally restricted melatonin secretion could be one internal zeitgeber that putatively distributes the endogenous circadian rhythmicity to all tissues expressing melatonin receptors.


Subject(s)
RNA, Messenger/biosynthesis , Receptors, Cell Surface/biosynthesis , Receptors, Cytoplasmic and Nuclear/biosynthesis , Actins/biosynthesis , Actins/genetics , Animals , Antisense Elements (Genetics) , Autoradiography , DNA, Complementary/biosynthesis , DNA, Complementary/isolation & purification , Male , Rats , Rats, Wistar , Receptors, Melatonin , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
11.
PLoS One ; 9(1): e87785, 2014.
Article in English | MEDLINE | ID: mdl-24489965

ABSTRACT

BACKGROUND: Electrical stimulation of the vagus nerve suppresses intestinal inflammation and normalizes gut motility in a mouse model of postoperative ileus. The exact anatomical interaction between the vagus nerve and the intestinal immune system remains however a matter of debate. In the present study, we provide additional evidence on the direct and indirect vagal innervation of the spleen and analyzed the anatomical evidence for neuroimmune modulation of macrophages by vagal preganglionic and enteric postganglionic nerve fibers within the intestine. METHODS: Dextran conjugates were used to label vagal preganglionic (motor) fibers projecting to the small intestine and spleen. Moreover, identification of the neurochemical phenotype of the vagal efferent fibers and enteric neurons was performed by immunofluorescent labeling. F4/80 antibody was used to label resident macrophages. RESULTS: Our anterograde tracing experiments did not reveal dextran-labeled vagal fibers or terminals in the mesenteric ganglion or spleen. Vagal efferent fibers were confined within the myenteric plexus region of the small intestine and mainly endings around nNOS, VIP and ChAT positive enteric neurons. nNOS, VIP and ChAT positive fibers were found in close proximity of intestinal resident macrophages carrying α7 nicotinic receptors. Of note, VIP receptors were found on resident macrophages located in close proximity of VIP positive nerve fibers. CONCLUSION: In the present study, we show that the vagus nerve does not directly interact with resident macrophages in the gut or spleen. Instead, the vagus nerve preferentially interacts with nNOS, VIP and ChAT enteric neurons located within the gut muscularis with nerve endings in close proximity of the resident macrophages.


Subject(s)
Intestine, Small/innervation , Macrophages/physiology , Spleen/innervation , Vagus Nerve/physiology , Acetylcholine/metabolism , Animals , Efferent Pathways , Female , Intestine, Small/cytology , Intestine, Small/metabolism , Lymph Nodes/cytology , Mice , Mice, Inbred BALB C , Myenteric Plexus/cytology , Myenteric Plexus/physiology , Neck , Nerve Growth Factors/metabolism , Nitric Oxide Synthase Type I/metabolism , Spleen/cytology , alpha7 Nicotinic Acetylcholine Receptor/metabolism
12.
PLoS One ; 9(7): e102211, 2014.
Article in English | MEDLINE | ID: mdl-25010202

ABSTRACT

BACKGROUND: Postoperative ileus is characterized by a transient impairment of the gastrointestinal motility after abdominal surgery. The intestinal inflammation, triggered by handling of the intestine, is the main factor responsible for the prolonged dysmotility of the gastrointestinal tract. Secondary lymphoid organs of the intestine were identified as essential components in the dissemination of inflammation to the entire gastrointestinal tract also called field effect. The involvement of the spleen, however, remains unclear. AIM: In this study, we investigated whether the spleen responds to manipulation of the intestine and participates in the intestinal inflammation underlying postoperative ileus. METHODS: Mice underwent Laparotomy (L) or Laparotomy followed by Intestinal Manipulation (IM). Twenty-four hours later, intestinal and colonic inflammation was assessed by QPCR and measurement of the intestinal transit was performed. Analysis of homeostatic chemokines in the spleen was performed by QPCR and splenic cell populations analysed by Flow Cytometry. Blockade of the egress of cells from the spleen was performed by administration of the Sphingosine-1-phosphate receptor 1 (S1P1) agonist CYM-5442 10 h after L/IM. RESULTS: A significant decrease in splenic weight and cellularity was observed in IM mice 24 h post-surgery, a phenomenon associated with a decreased splenic expression level of the homeostatic chemokine CCL19. Splenic denervation restored the expression of CCL19 and partially prevented the reduction of splenocytes in IM mice. Treatment with CYM-5442 prevented the egress of splenocytes but did not ameliorate the intestinal inflammation underlying postoperative ileus. CONCLUSIONS: Intestinal manipulation results in two distinct phenomena: local intestinal inflammation and a decrease in splenic cellularity. The splenic response relies on an alteration of cell trafficking in the spleen and is partially regulated by the splenic nerve. The spleen however does not participate in the intestinal inflammation during POI.


Subject(s)
Ileus/surgery , Inflammation/metabolism , Intestines/surgery , Spleen/metabolism , Animals , Disease Models, Animal , Gastrointestinal Motility/drug effects , Humans , Ileus/physiopathology , Indans/administration & dosage , Inflammation/pathology , Inflammation/surgery , Intestinal Mucosa/metabolism , Intestines/physiopathology , Male , Mice , Oxadiazoles/administration & dosage , Postoperative Complications/metabolism , Postoperative Complications/pathology , Postoperative Period , Receptors, Lysosphingolipid/antagonists & inhibitors , Spleen/drug effects
13.
Organogenesis ; 9(3): 216-23, 2013.
Article in English | MEDLINE | ID: mdl-23867810

ABSTRACT

Key physiological functions of the intestine are governed by nerves and neurotransmitters. This complex control relies on two neuronal systems: an extrinsic innervation supplied by the two branches of the autonomic nervous system and an intrinsic innervation provided by the enteric nervous system. As a result of constant exposure to commensal and pathogenic microflora, the intestine developed a tightly regulated immune system. In this review, we cover the current knowledge on the interactions between the gut innervation and the intestinal immune system. The relations between extrinsic and intrinsic neuronal inputs are highlighted with regards to the intestinal immune response. Moreover, we discuss the latest findings on mechanisms underlying inflammatory neural reflexes and examine their relevance in the context of the intestinal inflammation. Finally, we discuss some of the recent data on the identification of the gut microbiota as an emerging player influencing the brain function.


Subject(s)
Enteric Nervous System , Intestines , Animals , Humans , Intestines/innervation , Intestines/physiology , Mice , Microbiota , Neuroimmunomodulation , Spleen/innervation
14.
Chronobiol Int ; 29(7): 835-49, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22823867

ABSTRACT

The hypothalamic suprachiasmatic nucleus (SCN) is an essential component of the circadian timing system, and an important determinant of neuroendocrine and metabolic regulation. Recent data indicate a modulatory role for the immune system on the circadian timing system. The authors investigated how the circadian timing system affects the hypothalamo-pituitary-adrenal (HPA) axis and glucose regulatory responses evoked by an immune challenge induced by lipopolysaccharide (LPS). LPS-induced increases in corticosterone were minimal during the trough of the daily corticosterone rhythm; in contrast, LPS effects on glucose, glucagon, and insulin did not vary across time-of-day. Complete ablation of the SCN resulted in increased corticosterone responses but did not affect LPS-induced hyperglycemia. The paraventricular nucleus (PVN) of the hypothalamus is an important neuroendocrine and autonomic output pathway for hypothalamic information, as well as one of the main target areas of the SCN. Silencing the neuronal activity in the PVN did not affect the LPS-induced corticosterone surge and only slightly delayed the LPS-induced plasma glucose and glucagon responses. Finally, surgical interruption of the neuronal connection between hypothalamus and liver did not affect the corticosterone response but slightly delayed the LPS-induced glucose response. Together, these data support the previously proposed circadian modulation of LPS-induced neuroendocrine responses, but they are at variance with the suggested major role for the hypothalamic pacemaker on the autonomic output of the hypothalamus, as reflected by the effects of LPS on glucose homeostasis. The latter effects are more likely due to direct interactions of LPS with peripheral tissues, such as the liver.


Subject(s)
Blood Glucose/metabolism , Circadian Rhythm/physiology , Corticosterone/blood , Suprachiasmatic Nucleus/physiology , Animals , Circadian Rhythm/drug effects , Circadian Rhythm/immunology , Food Deprivation , Glucagon/blood , Hypothalamo-Hypophyseal System/physiology , Lipopolysaccharides/administration & dosage , Liver/innervation , Liver/physiology , Male , Paraventricular Hypothalamic Nucleus/physiology , Pituitary-Adrenal System/physiology , Rats , Rats, Wistar , Suprachiasmatic Nucleus/drug effects , Suprachiasmatic Nucleus/immunology , Sympathectomy, Chemical
15.
Life Sci ; 91(21-22): 1038-42, 2012 Nov 27.
Article in English | MEDLINE | ID: mdl-22580288

ABSTRACT

The gut immune system shares many signalling molecules and receptors with the autonomic nervous system. A good example is the vagal neurotransmitter acetylcholine (ACh), for which many immune cell types express cholinergic receptors (AChR). In the last decade the vagal nerve has emerged as an integral part of an immune regulation network via its release of ACh; a system coined "the cholinergic anti-inflammatory reflex". The perspective of cholinergic immune regulation in the gut mucosa has been widened by the recent discovery of populations of ACh producing immune cells in the spleen and other organs. As such, ACh, classically referred to as neurotransmitter, may serve a much broader function as bi-directional signalling molecule between neurons and non-neuronal cell types of the immune system.


Subject(s)
Acetylcholine/immunology , Immunity , Intestines/immunology , Receptors, Cholinergic/immunology , Animals , Humans , Signal Transduction
16.
Nat Rev Gastroenterol Hepatol ; 9(11): 675-83, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22801725

ABSTRACT

Patients undergoing an abdominal surgical procedure develop a transient episode of impaired gastrointestinal motility or postoperative ileus. Importantly, postoperative ileus is a major determinant of recovery after intestinal surgery and leads to increased morbidity and prolonged hospitalization, which is a great economic burden to health-care systems. Although a variety of strategies reduce postoperative ileus, including multimodal postoperative rehabilitation (fast-track care) and minimally invasive surgery, none of these methods have been completely successful in shortening the duration of postoperative ileus. The aetiology of postoperative ileus is multifactorial, but insights into the pathogenesis of postoperative ileus have identified intestinal inflammation, triggered by surgical handling, as the main mechanism. The importance of this inflammatory response in postoperative ileus is underscored by the beneficial effect of pharmacological interventions that block the influx of leukocytes. New insights into the pathophysiology of postoperative ileus and the involvement of the innate and the adaptive (T-helper type 1 cell-mediated immune response) immune system offer interesting and important new approaches to prevent postoperative ileus. In this Review, we discuss the latest insights into the mechanisms behind postoperative ileus and highlight new strategies to intervene in the postoperative inflammatory cascade.


Subject(s)
Ileus/drug therapy , Ileus/physiopathology , Postoperative Complications/drug therapy , Postoperative Complications/physiopathology , Adaptive Immunity/physiology , Ghrelin/agonists , Humans , Immunity, Innate/physiology , Inflammation/physiopathology , Naphthoquinones/therapeutic use , Serotonin 5-HT4 Receptor Agonists/therapeutic use
17.
Article in English | MEDLINE | ID: mdl-23320135

ABSTRACT

INTRODUCTION: Postoperative ileus (POI) is characterized by a transient inhibition of coordinated motility of the gastrointestinal (GI) tract after abdominal surgery and leads to increased morbidity and prolonged hospitalization. Currently, intestinal manipulation of the intestine is widely used as a preclinical model of POI. The technique used to manipulate the intestine is however highly variable and difficult to standardize, leading to large variations and inconsistent findings between different investigators. Therefore, we developed a device by which a fixed and adjustable pressure can be applied during intestinal manipulation. METHODS: The standardized pressure manipulation method was developed using the purpose-designed device. First, the effect of graded manipulation was examined on postoperative GI transit. Next, this new technique was compared to the conventional manipulation technique used in previous studies. GI transit was measured by evaluating the intestinal distribution of orally gavaged fluorescein isothiocyanate (FITC)-labeled dextran. Infiltration of myeloperoxidase positive cells and cytokine production (ELISA) in the muscularis externa of the intestine were assessed. RESULTS: Increasing pressures resulted in a graded reduction of intestinal transit and was associated with intestinal inflammation as demonstrated by influx of leukocytes and increased levels of IL-6, IL-1ß and MCP-1 compared to control mice. With an applied pressure of 9 grams a similar delay in intestinal transit could be obtained with a smaller standard deviation, leading to a reduced intra-individual variation. CONCLUSIONS: This method provides a reproducible model with small variation to study the pathophysiology of POI and to evaluate new anti-inflammatory strategies.

18.
Essays Biochem ; 49(1): 137-51, 2011 Jun 30.
Article in English | MEDLINE | ID: mdl-21819389

ABSTRACT

In mammals many behaviours (e.g. sleep-wake, feeding) as well as physiological (e.g. body temperature, blood pressure) and endocrine (e.g. plasma corticosterone concentration) events display a 24 h rhythmicity. These 24 h rhythms are induced by a timing system that is composed of central and peripheral clocks. The highly co-ordinated output of the hypothalamic biological clock not only controls the daily rhythm in sleep-wake (or feeding-fasting) behaviour, but also exerts a direct control over many aspects of hormone release and energy metabolism. First, we present the anatomical connections used by the mammalian biological clock to enforce its endogenous rhythmicity on the rest of the body, especially the neuro-endocrine and energy homoeostatic systems. Subsequently, we review a number of physiological experiments investigating the functional significance of this neuro-anatomical substrate. Together, this overview of experimental data reveals a highly specialized organization of connections between the hypothalamic pacemaker and neuro-endocrine system as well as the pre-sympathetic and pre-parasympathetic branches of the autonomic nervous system.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , Corticosterone/metabolism , Hypothalamus/physiology , Suprachiasmatic Nucleus/physiology , Animals , Autonomic Nervous System/physiology , Corticosterone/physiology , Endocrine System/metabolism , Endocrine System/physiology , Humans , Neurons/physiology , Suprachiasmatic Nucleus/metabolism
19.
PLoS One ; 4(5): e5650, 2009 May 21.
Article in English | MEDLINE | ID: mdl-19478857

ABSTRACT

BACKGROUND: The biological clock, located in the hypothalamic suprachiasmatic nucleus (SCN), controls the daily rhythms in physiology and behavior. Early studies demonstrated that light exposure not only affects the phase of the SCN but also the functional activity of peripheral organs. More recently it was shown that the same light stimulus induces immediate changes in clock gene expression in the pineal and adrenal, suggesting a role of peripheral clocks in the organ-specific output. In the present study, we further investigated the immediate effect of nocturnal light exposure on clock genes and metabolism-related genes in different organs of the rat. In addition, we investigated the role of the autonomic nervous system as a possible output pathway of the SCN to modify the activity of the liver after light exposure. METHODOLOGY AND PRINCIPAL FINDINGS: First, we demonstrated that light, applied at different circadian times, affects clock gene expression in a different manner, depending on the time of day and the organ. However, the changes in clock gene expression did not correlate in a consistent manner with those of the output genes (i.e., genes involved in the functional output of an organ). Then, by selectively removing the autonomic innervation to the liver, we demonstrated that light affects liver gene expression not only via the hormonal pathway but also via the autonomic input. CONCLUSION: Nocturnal light immediately affects peripheral clock gene expression but without a clear correlation with organ-specific output genes, raising the question whether the peripheral clock plays a "decisive" role in the immediate (functional) response of an organ to nocturnal light exposure. Interestingly, the autonomic innervation of the liver is essential to transmit the light information from the SCN, indicating that the autonomic nervous system is an important gateway for the SCN to cause an immediate resetting of peripheral physiology after phase-shift inducing light exposures.


Subject(s)
Autonomic Nervous System/radiation effects , Biological Clocks/genetics , Biological Clocks/radiation effects , Darkness , Gene Expression Regulation/radiation effects , Liver/innervation , Organ Specificity/genetics , Adrenal Glands/metabolism , Adrenal Glands/radiation effects , Animals , Autonomic Denervation , Circadian Rhythm/genetics , Circadian Rhythm/radiation effects , Hormones/metabolism , Liver/metabolism , Liver/radiation effects , Male , Organ Specificity/radiation effects , Pineal Gland/metabolism , Pineal Gland/radiation effects , Rats , Rats, Wistar
20.
Biochem Biophys Res Commun ; 330(1): 18-26, 2005 Apr 29.
Article in English | MEDLINE | ID: mdl-15781226

ABSTRACT

The circadian expression patterns of genes encoding for proteins that make up the core of the circadian clock were measured in rat retina using real-time quantitative PCR (qPCR). Transcript levels of several genes previously used for normalization of qPCR assays were determined and the effect of ischemia-reperfusion on the expression of clock genes was studied. Statistically significant circadian changes in transcript levels were found for: Per2, Per3, Cry2, Bmal1, Rora, Rorb, and Rorc with changes ranging between 1.6- and 2.6-fold. No changes were found for Per1, Cry1, Clock, Rev-erb alpha, and Rev-erb beta. Significant differences in transcript levels were observed for several candidate reference genes: HPRT, GAPDH, rhodopsin, and Thy1 and, consequently, the use of these genes for normalization purposes in qPCR or Northern blots may lead to erroneous conclusions. Ischemia-reperfusion leads to a persistent decrease of Per1 and Cry2, which may be related to the selective degeneration of amacrine and ganglion cells. We conclude that while all clock genes are expressed in the retina, only a few show a clear circadian pattern.


Subject(s)
Circadian Rhythm , Retina/metabolism , Trans-Activators/genetics , Animals , Base Sequence , Blotting, Northern , CLOCK Proteins , DNA Primers , Electrophoresis, Agar Gel , Male , Polymerase Chain Reaction , RNA, Messenger/genetics , Rats , Rats, Wistar , Reperfusion Injury/genetics
SELECTION OF CITATIONS
SEARCH DETAIL