71 results on '"De Lartigue G"'
Search Results
2. Vagal afferent nerve ghrelin signaling influences energy balance and episodic memory
- Author
-
Davis, EA, primary, Suarez, AN, additional, Liu, CM, additional, Cortella, AM, additional, de Lartigue, G, additional, and Kanoski, SE, additional
- Published
- 2020
- Full Text
- View/download PDF
3. Role of gut to brain signaling in the development of diet-induced obesity
- Author
-
de Lartigue, G., primary
- Published
- 2015
- Full Text
- View/download PDF
4. Chronic administration of low dose bacterial lipopolysaccharide (LPS) inhibits cholecystokinin (CCK)-induced satiation
- Author
-
Barbier De La Serre, C., primary, De Lartigue, G., additional, Sharp, J., additional, and Raybould, H., additional
- Published
- 2011
- Full Text
- View/download PDF
5. Neuropeptide transmitters released from VAN into the NTS play an important role in regulating food intake
- Author
-
De Lartigue, G., primary, Darling, R.A., additional, and Raybould, H.E., additional
- Published
- 2011
- Full Text
- View/download PDF
6. Cholecystokinin Regulates Expression of Y2 Receptors in Vagal Afferent Neurons Serving the Stomach
- Author
-
Burdyga, G., primary, de Lartigue, G., additional, Raybould, H. E., additional, Morris, R., additional, Dimaline, R., additional, Varro, A., additional, Thompson, D. G., additional, and Dockray, G. J., additional
- Published
- 2008
- Full Text
- View/download PDF
7. Cocaine- and Amphetamine-Regulated Transcript: Stimulation of Expression in Rat Vagal Afferent Neurons by Cholecystokinin and Suppression by Ghrelin
- Author
-
de Lartigue, G., primary, Dimaline, R., additional, Varro, A., additional, and Dockray, G. J., additional
- Published
- 2007
- Full Text
- View/download PDF
8. Bifidobacteria isolated from infants and cultured on human milk oligosaccharides affect intestinal epithelial function.
- Author
-
Chichlowski M, De Lartigue G, German JB, Raybould HE, Mills DA, Chichlowski, Maciej, De Lartigue, Guillaume, German, J Bruce, Raybould, Helen E, and Mills, David A
- Published
- 2012
- Full Text
- View/download PDF
9. Hepatic vagal afferents convey clock-dependent signals to regulate circadian food intake.
- Author
-
Woodie LN, Melink LC, Midha M, de Araújo AM, Geisler CE, Alberto AJ, Krusen BM, Zundell DM, de Lartigue G, Hayes MR, and Lazar MA
- Subjects
- Animals, Male, Mice, Brain physiology, Circadian Clocks, CLOCK Proteins metabolism, CLOCK Proteins genetics, Homeostasis, Mice, Inbred C57BL, Vagotomy, Weight Gain, Afferent Pathways physiology, Circadian Rhythm physiology, Diet, High-Fat adverse effects, Eating physiology, Liver innervation, Obesity physiopathology, Vagus Nerve physiopathology, Vagus Nerve surgery
- Abstract
Circadian desynchrony induced by shiftwork or jet lag is detrimental to metabolic health, but how synchronous or desynchronous signals are transmitted among tissues is unknown. We report that liver molecular clock dysfunction is signaled to the brain through the hepatic vagal afferent nerve (HVAN), leading to altered food intake patterns that are corrected by ablation of the HVAN. Hepatic branch vagotomy also prevents food intake disruptions induced by high-fat diet feeding and reduces body weight gain. Our findings reveal a homeostatic feedback signal that relies on communication between the liver and the brain to control circadian food intake patterns. This identifies the hepatic vagus nerve as a potential therapeutic target for obesity in the setting of chronodisruption.
- Published
- 2024
- Full Text
- View/download PDF
10. Intestinal serotonergic vagal signaling as a mediator of microbiota-induced hypertension.
- Author
-
de Araujo A, Sree Kumar H, Yang T, Plata AA, Dirr EW, Bearss N, Baekey DM, Miller DS, Donertas-Ayaz B, Ahmari N, Singh A, Kalinoski AL, Garrett TJ, Martyniuk CJ, de Lartigue G, and Zubcevic J
- Abstract
Hypertension is a pervasive global health challenge, impacting over a billion individuals worldwide. Despite strides in therapeutic strategies, a significant proportion of patients remain resistant to the currently available therapies. While conventional treatments predominantly focus on cardiac, renal, and cerebral targets, emerging research underscores the pivotal role of the gut and its microbiota. Yet, the precise mechanisms governing interactions between the gut microbiota and the host blood pressure remain unclear. Here we describe a neural host-microbiota interaction that is mediated by the intestinal serotonin (5-HT) signaling via vagal 5HT3a receptors and which is crucial for maintenance of blood pressure homeostasis. Notably, a marked decrease in both intestinal 5-HT and vagal 5HT3aR signaling is observed in hypertensive rats, and in rats subjected to fecal microbiota transplantation from hypertensive rats. Leveraging an intersectional genetic strategy in a Cre rat line, we demonstrate that intestinal 5HT3aR vagal signaling is a crucial link between the gut microbiota and blood pressure homeostasis and that recovery of 5-HT signaling in colon innervating vagal neurons can alleviate hypertension. This paradigm-shifting finding enhances our comprehension of hypertensive pathophysiology and unveils a promising new therapeutic target for combating resistant hypertension associated with gut dysbiosis.
- Published
- 2024
- Full Text
- View/download PDF
11. Mitragynine and morphine produce dose-dependent bimodal action on food but not water intake in rats.
- Author
-
Zuarth Gonzalez JD, Mottinelli M, McCurdy CR, de Lartigue G, McMahon LR, and Wilkerson JL
- Subjects
- Animals, Male, Rats, Drinking drug effects, Narcotic Antagonists pharmacology, Naltrexone pharmacology, Naltrexone administration & dosage, Analgesics, Opioid pharmacology, Analgesics, Opioid administration & dosage, Feeding Behavior drug effects, Secologanin Tryptamine Alkaloids pharmacology, Secologanin Tryptamine Alkaloids administration & dosage, Morphine pharmacology, Morphine administration & dosage, Rats, Sprague-Dawley, Dose-Response Relationship, Drug, Eating drug effects
- Abstract
Kratom ( Mitragyna speciosa ), containing the primary alkaloid mitragynine, has emerged as an alternative self-treatment for opioid use disorder. Mitragynine binds numerous receptor types, including opioid receptors, which are known to modulate food consumption. However, the ability of acute mitragynine to modulate food consumption remains unknown. The current study assessed the effects of acute mitragynine or morphine administration on unconditioned food and water intake in 16 Sprague-Dawley rats. Food and water intake changes were monitored in response to morphine, mitragynine (1.78-56 mg/kg ip), saline, or vehicle controls for 12 h, starting at the onset of the dark cycle. Naltrexone pretreatment was used to examine pharmacological specificity. Both morphine and mitragynine demonstrated a biphasic food intake dose-effect, with low doses (5.6 mg/kg) increasing and high doses (56 mg/kg) decreasing food intake. All morphine doses reduced water intake; however, only the highest dose of mitragynine (56 mg/kg) reduced water intake. Naltrexone attenuated both stimulatory and inhibitory effects of morphine on food intake, but only the stimulatory effect of mitragynine. In conclusion, low doses of mitragynine stimulate food intake via opioid-related pathways, while high doses likely recruit other targets. NEW & NOTEWORTHY This study reveals that morphine and the kratom alkaloid mitragynine produce dose-dependent effects on feeding in rats. Low doses stimulate food intake via opioid pathways, while high doses decrease consumption through nonopioid mechanisms. Morphine potently suppresses water intake at all doses, whereas only high doses of mitragynine reduce drinking. These findings provide novel insights into the complex opioid and nonopioid mechanisms underlying the effects of mitragynine on ingestive behaviors.
- Published
- 2024
- Full Text
- View/download PDF
12. Separate gut-brain circuits for fat and sugar reinforcement combine to promote overeating.
- Author
-
McDougle M, de Araujo A, Singh A, Yang M, Braga I, Paille V, Mendez-Hernandez R, Vergara M, Woodie LN, Gour A, Sharma A, Urs N, Warren B, and de Lartigue G
- Published
- 2024
- Full Text
- View/download PDF
13. Transfer with microbiota from lean donors prevents excessive weight gain and restores gut-brain vagal signaling in obese rats maintained on a high fat diet.
- Author
-
Minaya DM, Kim JS, Kirkland R, Allen J, Cullinan S, Maclang N, de Lartigue G, and de La Serre CB
- Abstract
Background: The collection of microorganisms, mainly bacteria, which live in the gastrointestinal (GI) tract are collectible known as the gut microbiota. GI bacteria play an active role in regulation of the host's immune system and metabolism, as well as certain pathophysiological processes. Diet is the main factor modulating GI microbiota composition and recent studies have shown that high fat (HF) diets induce detrimental changes, known as dysbiosis, in the GI bacterial makeup. HF diet induced microbiota dysbiosis has been associated with structural and functional changes in gut-brain vagally mediated signaling system, associated with overeating and obesity. Although HF-driven changes in microbiota composition are sufficient to alter vagal signaling, it is unknown if restoring normal microbiota in obesity can improve gut-brain signaling and metabolic outcomes. In this study, we evaluated the effect of lean gut microbiota transfer in obese, vagally compromised, rats on gut-brain communication, food intake, and body weight. Male Sprague-Dawley rats were maintained on regular chow, or 45% HF diet for nine weeks followed by three weeks of microbiota depletion using an antibiotic cocktail. The animals were then divided into four groups (n=10 each): LF - control group on regular chow, LF-LF - chow fed animals that received antibiotics and microbiota from chow fed animals, HF-LF - HF fed animals that received microbiota from chow fed animals, and HF-HF - HF fed animals that received microbiota from HF fed animals. Animals were gavaged with donor microbiota for three consecutive days on week one and once a week thereafter for three more weeks. HF-LF animals received inulin as a prebiotic to aid the establishment of the lean microbiome., Results: We found that transferring a LF microbiota to HF fed animals (HF-LF) reduced caloric intake during the light phase when compared with HF-HF rats and prevented additional excessive weight gain. We did not observe significant changes in the density of vagal afferents terminating in the brainstem among the groups, however, HF-LF animals displayed an increase in postprandial activation of both primary sensory neurons innervating the GI tract and brainstem secondary neurons., Conclusions: We concluded from these data that normalizing microbiota composition in obese rats improves gut-brain communication and restores normal feeding patterns which was associated with a reduction in weight gain., Competing Interests: Competing interests The authors declare that they have no competing interests.
- Published
- 2024
- Full Text
- View/download PDF
14. Alleviating Hypertension by Selectively Targeting Angiotensin Receptor-Expressing Vagal Sensory Neurons.
- Author
-
Baumer-Harrison C, Elsaafien K, Johnson DN, Peñaloza Aponte JD, de Araujo A, Patel S, Bruce EB, Harden SW, Frazier CJ, Scott KA, de Lartigue G, Krause EG, and de Kloet AD
- Subjects
- Mice, Male, Female, Animals, Solitary Nucleus physiology, Sensory Receptor Cells, Blood Pressure physiology, Phenylephrine pharmacology, Ion Channels, Desoxycorticosterone Acetate pharmacology, Hypertension, Red Fluorescent Protein
- Abstract
Cardiovascular homeostasis is maintained, in part, by neural signals arising from arterial baroreceptors that apprise the brain of blood volume and pressure. Here, we test whether neurons within the nodose ganglia that express angiotensin type-1a receptors (referred to as NG
AT1aR ) serve as baroreceptors that differentially influence blood pressure (BP) in male and female mice. Using Agtr1a -Cre mice and Cre-dependent AAVs to direct tdTomato to NGAT1aR , neuroanatomical studies revealed that NGAT1aR receive input from the aortic arch, project to the caudal nucleus of the solitary tract (NTS), and synthesize mechanosensitive ion channels, Piezo1/2 To evaluate the functionality of NGAT1aR , we directed the fluorescent calcium indicator (GCaMP6s) or the light-sensitive channelrhodopsin-2 (ChR2) to Agtr1a -containing neurons. Two-photon intravital imaging in Agtr1a -GCaMP6s mice revealed that NGAT1aR couple their firing to elevated BP, induced by phenylephrine (i.v.). Furthermore, optical excitation of NGAT1aR at their soma or axon terminals within the caudal NTS of Agtr1a -ChR2 mice elicited robust frequency-dependent decreases in BP and heart rate, indicating that NGAT1aR are sufficient to elicit appropriate compensatory responses to vascular mechanosensation. Optical excitation also elicited hypotensive and bradycardic responses in ChR2-expressing mice that were subjected to deoxycorticosterone acetate (DOCA)-salt hypertension; however, the duration of these effects was altered, suggestive of hypertension-induced impairment of the baroreflex. Similarly, increased GCaMP6s fluorescence observed after administration of phenylephrine was delayed in mice subjected to DOCA-salt or chronic delivery of angiotensin II. Collectively, these results reveal the structure and function of NGAT1aR and suggest that such neurons may be exploited to discern and relieve hypertension., Competing Interests: The authors declare no competing financial interests., (Copyright © 2024 the authors.)- Published
- 2024
- Full Text
- View/download PDF
15. Transfer of microbiota from lean donors in combination with prebiotics prevents excessive weight gain and improves gut-brain vagal signaling in obese rats.
- Author
-
Minaya DM, Kim JS, Kirkland R, Allen J, Cullinan S, Maclang N, de Lartigue G, and de La Serre C
- Subjects
- Animals, Male, Rats, Dysbiosis microbiology, Rats, Sprague-Dawley, Bacteria classification, Bacteria isolation & purification, Bacteria genetics, Bacteria metabolism, Fecal Microbiota Transplantation, Brain metabolism, Signal Transduction, Obesity microbiology, Obesity metabolism, Gastrointestinal Microbiome drug effects, Prebiotics administration & dosage, Brain-Gut Axis physiology, Diet, High-Fat adverse effects, Vagus Nerve, Weight Gain drug effects
- Abstract
Gastrointestinal (GI) microbiota plays an active role in regulating the host's immune system and metabolism, as well as certain pathophysiological processes. Diet is the main factor modulating GI microbiota composition and studies have shown that high fat (HF) diets induce detrimental changes (dysbiosis) in the GI bacterial makeup. HF diet induced dysbiosis has been associated with structural and functional changes in gut-brain vagally mediated signaling system, associated with overeating and obesity. Although HF-driven changes in microbiota composition are sufficient to alter vagal signaling, it is unknown if improving microbiota composition after diet-induced obesity has been established can ameliorate gut-brain signaling and metabolic outcomes. In this study, we evaluated the effect of lean gut microbiota transfer in obese, vagally compromised, rats on gut-brain communication, food intake, and body weight. Male rats were maintained on regular chow or 45% HF diet for nine weeks followed by three weeks of microbiota depletion using antibiotics. The animals were then divided into four groups ( n = 10 each): LF - control fed regular chow, LF-LF - chow fed animals that received microbiota from chow fed donors, HF-LF - HF fed animals that received microbiota from chow fed donors, and HF-HF - HF fed animals that received microbiota from HF fed donors. HF-LF animals received inulin as a prebiotic to aid the establishment of the lean microbiome. We found that transferring a LF microbiota to HF fed animals (HF-LF) reduced caloric intake during the light phase when compared with HF-HF rats and prevented additional excessive weight gain. HF-LF animals displayed an increase in postprandial activation of both primary sensory neurons innervating the GI tract and brainstem secondary neurons. We concluded from these data that improving microbiota composition in obese rats is sufficient to ameliorate gut-brain communication and restore normal feeding patterns which was associated with a reduction in weight gain.
- Published
- 2024
- Full Text
- View/download PDF
16. Hepatic Vagal Afferents Convey Clock-Dependent Signals to Regulate Circadian Food Intake.
- Author
-
Woodie LN, Melink LC, Midha M, de Araújo AM, Geisler CE, Alberto AJ, Krusen BM, Zundell DM, de Lartigue G, Hayes MR, and Lazar MA
- Abstract
Circadian desynchrony induced by shiftwork or jetlag is detrimental to metabolic health, but how synchronous/desynchronous signals are transmitted among tissues is unknown. Here we report that liver molecular clock dysfunction is signaled to the brain via the hepatic vagal afferent nerve (HVAN), leading to altered food intake patterns that are corrected by ablation of the HVAN. Hepatic branch vagotomy also prevents food intake disruptions induced by high-fat diet feeding and reduces body weight gain. Our findings reveal a previously unrecognized homeostatic feedback signal that relies on synchrony between the liver and the brain to control circadian food intake patterns. This identifies the hepatic vagus nerve as a therapeutic target for obesity in the setting of chrono-disruption., One Sentence Summary: The hepatic vagal afferent nerve signals internal circadian desynchrony between the brain and liver to induce maladaptive food intake patterns.
- Published
- 2023
- Full Text
- View/download PDF
17. Ventral hippocampus neurons encode meal-related memory.
- Author
-
Décarie-Spain L, Gu C, Lauer LT, Subramanian KS, Chehimi SN, Kao AE, Deng I, Bashaw AG, Klug ME, Galbokke AH, Donohue KN, Yang M, de Lartigue G, Myers KP, Crist RC, Reiner BC, Hayes MR, and Kanoski SE
- Abstract
The ability to encode and retrieve meal-related information is critical to efficiently guide energy acquisition and consumption, yet the underlying neural processes remain elusive. Here we reveal that ventral hippocampus (HPCv) neuronal activity dynamically elevates during meal consumption and this response is highly predictive of subsequent performance in a foraging-related spatial memory task. Targeted recombination-mediated ablation of HPCv meal-responsive neurons impairs foraging-related spatial memory without influencing food motivation, anxiety-like behavior, or escape-mediated spatial memory. These HPCv meal-responsive neurons project to the lateral hypothalamic area (LHA) and single-nucleus RNA sequencing and in situ hybridization analyses indicate they are enriched in serotonin 2a receptors (5HT2aR). Either chemogenetic silencing of HPCv-to-LHA projections or intra-HPCv 5HT2aR antagonist yielded foraging-related spatial memory deficits, as well as alterations in caloric intake and the temporal sequence of spontaneous meal consumption. Collective results identify a population of HPCv neurons that dynamically respond to eating to encode meal-related memories., Competing Interests: CONFLICT OF INTEREST BCR and MRH both receive research funding from Novo Nordisk and Boehringer Ingelheim that was not used in support of these studies. MRH receives research funding from Eli Lilly & Co., Gila Therapeutics, and Pfizer that was not used in support of these studies. MRH is CEO of Cantius Therapeutics, LLC which pursues biological work unrelated to the current study. All other authors declare no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
18. Separate orexigenic hippocampal ensembles shape dietary choice by enhancing contextual memory and motivation.
- Author
-
Yang M, Singh A, McDougle M, Décarie-Spain L, Kanoski S, and de Lartigue G
- Abstract
The hippocampus (HPC), traditionally known for its role in learning and memory, has emerged as a controller of food intake. While prior studies primarily associated the HPC with food intake inhibition, recent research suggests a critical role in appetitive processes. We hypothesized that orexigenic HPC neurons differentially respond to fats and/or sugars, potent natural reinforcers that contribute to obesity development. Results uncover previously-unrecognized, spatially-distinct neuronal ensembles within the dorsal HPC (dHPC) that are responsive to separate nutrient signals originating from the gut. Using activity-dependent genetic capture of nutrient-responsive HPC neurons, we demonstrate a causal role of both populations in promoting nutrient-specific preference through different mechanisms. Sugar-responsive neurons encode an appetitive spatial memory engram for meal location, whereas fat-responsive neurons selectively enhance the preference and motivation for fat intake. Collectively, these findings uncover a neural basis for the exquisite specificity in processing macronutrient signals from a meal that shape dietary choices.
- Published
- 2023
- Full Text
- View/download PDF
19. The gut-brain axis mediates bacterial driven modulation of reward signaling.
- Author
-
Kim JS, Williams KC, Kirkland RA, Schade R, Freeman KG, Cawthon CR, Rautmann AW, Smith JM, Edwards GL, Glenn TC, Holmes PV, de Lartigue G, and de La Serre CB
- Subjects
- Rats, Male, Animals, 3,4-Dihydroxyphenylacetic Acid, Reward, Bacteria, Brain-Gut Axis, Feeding Behavior physiology
- Abstract
Objective: Our goal is to investigate if microbiota composition modulates reward signaling and assess the role of the vagus in mediating microbiota to brain communication., Methods: Male germ-free Fisher rats were colonized with gastrointestinal contents from chow (low fat (LF) ConvLF) or HF (ConvHF) fed rats., Results: Following colonization, ConvHF rats consumed significantly more food than ConvLF animals. ConvHF rats displayed lower feeding-induced extracellular DOPAC levels (a metabolite of dopamine) in the Nucleus Accumbens (NAc) as well as reduced motivation for HF foods compared to ConvLF rats. Dopamine receptor 2 (DDR2) expression levels in the NAc were also significantly lower in ConvHF animals. Similar deficits were observed in conventionally raised HF fed rats, showing that diet-driven alteration in reward can be initiated via microbiota. Selective gut to brain deafferentation restored DOPAC levels, DRD2 expression, and motivational drive in ConvHF rats., Conclusions: We concluded from these data that a HF-type microbiota is sufficient to alter appetitive feeding behavior and that bacteria to reward communication is mediated by the vagus nerve., (Copyright © 2023 The Author(s). Published by Elsevier GmbH.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
20. Mechanosensation of the heart and gut elicits hypometabolism and vigilance in mice.
- Author
-
Scott KA, Tan Y, Johnson DN, Elsaafien K, Baumer-Harrison C, Eikenberry SA, Sa JM, de Lartigue G, de Kloet AD, and Krause EG
- Abstract
Interoception broadly refers to awareness of one's internal milieu. Vagal sensory afferents monitor the internal milieu and maintain homeostasis by engaging brain circuits that alter physiology and behavior. While the importance of the body-to-brain communication that underlies interoception is implicit, the vagal afferents and corresponding brain circuits that shape perception of the viscera are largely unknown. Here, we use mice to parse neural circuits subserving interoception of the heart and gut. We determine vagal sensory afferents expressing the oxytocin receptor, hereafter referred to as NDG
Oxtr , send projections to the aortic arch or stomach and duodenum with molecular and structural features indicative of mechanosensation. Chemogenetic excitation of NDGOxtr significantly decreases food and water consumption, and remarkably, produces a torpor-like phenotype characterized by reductions in cardiac output, body temperature, and energy expenditure. Chemogenetic excitation of NDGOxtr also creates patterns of brain activity associated with augmented hypothalamic-pituitary-adrenal axis activity and behavioral indices of vigilance. Recurrent excitation of NDGOxtr suppresses food intake and lowers body mass, indicating that mechanosensation of the heart and gut can exert enduring effects on energy balance. These findings suggest that the sensation of vascular stretch and gastrointestinal distention may have profound effects on whole body metabolism and mental health., Competing Interests: Competing interest declaration: The authors declare no competing interests.- Published
- 2023
- Full Text
- View/download PDF
21. Non-Nutritive Sweetened Beverages Impair Therapeutic Benefits of Metformin in Prediabetic Diet-Induced Obese Mice.
- Author
-
Singh A, Rourk K, Bernier A, and de Lartigue G
- Subjects
- Animals, Mice, Diet, High-Fat adverse effects, Mice, Obese, Obesity drug therapy, Obesity etiology, Saccharin, Glucose Intolerance, Diabetes Mellitus, Type 2 drug therapy, High Fructose Corn Syrup adverse effects, Metformin pharmacology, Metformin therapeutic use, Non-Nutritive Sweeteners adverse effects, Prediabetic State, Sugar-Sweetened Beverages
- Abstract
Metformin, a frontline therapy for type 2 diabetes and related metabolic diseases, results in variable outcomes. This study aimed to investigate whether sweetened beverages (caloric or non-caloric) affect the therapeutic benefits of metformin on glucose, food intake, and weight loss in diet-induced obesity. Mice were given a high-fat diet and sweetened water for 8 weeks to induce obesity and glucose intolerance. Then, mice were randomized to receive metformin in either water, high-fructose corn syrup (HFCS), or the non-nutritive sweetener saccharin for 6 weeks. After 6 weeks of metformin treatment, all groups had improved glucose tolerance compared to pretreatment. However, saccharin resulted in worse glucose tolerance and weight gain outcomes than the water or HFCS groups and correlated with lower plasma growth differentiation factor 15 levels. In conclusion, reducing non-nutritive sweetener consumption during metformin therapy is recommended to avoid impairing the therapeutic effects of metformin on body weight and glucose homeostasis.
- Published
- 2023
- Full Text
- View/download PDF
22. Asymmetric control of food intake by left and right vagal sensory neurons.
- Author
-
de Araujo AM, Braga I, Leme G, Singh A, McDougle M, Smith J, Vergara M, Yang M, Lin M, Khoshbouei H, Krause E, de Oliveira AG, and de Lartigue G
- Abstract
We investigated the lateralization of gut-innervating vagal sensory neurons and their roles in feeding behavior. Using genetic, anatomical, and behavioral analyses, we discovered a subset of highly lateralized vagal sensory neurons with distinct sensory responses to intestinal stimuli. Our results demonstrated that left vagal sensory neurons (LNG) are crucial for distension-induced satiety, while right vagal sensory neurons (RNG) mediate preference for nutritive foods. Furthermore, these lateralized neurons engage different central circuits, with LNG neurons recruiting brain regions associated with energy balance and RNG neurons activating areas related to salience, memory, and reward. Altogether, our findings unveil the diverse roles of asymmetrical gut-vagal-brain circuits in feeding behavior, offering new insights for potential therapeutic interventions targeting vagal nerve stimulation in metabolic and neuropsychiatric diseases., Competing Interests: Competing interests: The authors have nothing to report.
- Published
- 2023
- Full Text
- View/download PDF
23. MPYS Modulates Fatty Acid Metabolism and Immune Tolerance at Homeostasis Independent of Type I IFNs.
- Author
-
Mansouri S, Gogoi H, Patel S, Katikaneni DS, Singh A, Aybar-Torres A, de Lartigue G, and Jin L
- Subjects
- Adult, Animals, Humans, Mice, Homeostasis, Mice, Knockout, Interferon Type I, Fatty Acids metabolism, Immune Tolerance, Membrane Proteins metabolism
- Abstract
MPYS/STING (stimulator of IFN genes) senses cyclic dinucleotides (CDNs), generates type I IFNs, and plays a critical role in infection, inflammation, and cancer. In this study, analyzing genotype and haplotype data from the 1000 Genomes Project, we found that the R71H-G230A-R293Q (HAQ) MPYS allele frequency increased 57-fold in East Asians compared with sub-Saharan Africans. Meanwhile, the G230A-R293Q (AQ) allele frequency decreased by 98% in East Asians compared with sub-Saharan Africans. We propose that the HAQ and AQ alleles underwent a natural selection during the out-of-Africa migration. We used mouse models of HAQ and AQ to investigate the underlying mechanism. We found that the mice carrying the AQ allele, which disappeared in East Asians, had normal CDN-type I IFN responses. Adult AQ mice, however, had less fat mass than did HAQ or wild-type mice on a chow diet. AQ epididymal adipose tissue had increased regulatory T cells and M2 macrophages with protein expression associated with enhanced fatty acid oxidation. Conditional knockout mice and adoptive cell transfer indicate a macrophage and regulatory T cell-intrinsic role of MPYS in fatty acid metabolism. Mechanistically, AQ/IFNAR1-/- mice had a similar lean phenotype as for the AQ mice. MPYS intrinsic tryptophan fluorescence revealed that the R71H change increased MPYS hydrophilicity. Lastly, we found that the second transmembrane (TM) and the TM2-TM3 linker region of MPYS interact with activated fatty acid, fatty acyl-CoA. In summary, studying the evolution of the human MPYS gene revealed an MPYS function in modulating fatty acid metabolism that may be critical during the out-of-Africa migration., (Copyright © 2022 by The American Association of Immunologists, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
24. Neural Pathway for Gut Feelings: Vagal Interoceptive Feedback From the Gastrointestinal Tract Is a Critical Modulator of Anxiety-like Behavior.
- Author
-
Krieger JP, Asker M, van der Velden P, Börchers S, Richard JE, Maric I, Longo F, Singh A, de Lartigue G, and Skibicka KP
- Subjects
- Animals, Feedback, Female, Gastrointestinal Tract, Male, Neural Pathways physiology, Rats, Saporins metabolism, gamma-Aminobutyric Acid metabolism, Anxiety, Vagus Nerve metabolism
- Abstract
Background: Anxiety disorders are associated with an altered perception of the body's internal state. Therefore, understanding the neuronal basis of interoception can foster novel anxiety therapies. In rodents, the feeding status bidirectionally modulates anxiety-like behavior but how the sensing of gastrointestinal state affects anxiety remains unclear., Methods: We combined chemogenetics, neuropharmacology, and behavioral approaches in male and female rats to test whether vagal afferents terminating in the gastrointestinal tract mediate feeding-induced tuning of anxiety. Using saporin-based lesions and transcriptomics, we investigated the chronic impact of this gut-brain circuit on anxiety-like behavior., Results: Both feeding and selective chemogenetic activation of gut-innervating vagal afferents increased anxiety-like behavior. Conversely, chemogenetic inhibition blocked the increase in anxiety-like behavior induced by feeding. Using a selective saporin-based lesion, we demonstrate that the loss of gut-innervating vagal afferent signaling chronically reduces anxiety-like behavior in male rats but not in female rats. We next identify a vagal circuit that connects the gut to the central nucleus of the amygdala, using anterograde transsynaptic tracing from the nodose ganglia. Lesion of this gut-brain vagal circuit modulated the central amygdala transcriptome in both sexes but selectively affected a network of GABA (gamma-aminobutyric acid)-related genes only in males, suggesting a potentiation of inhibitory control. Blocking GABAergic signaling in the central amygdala re-established normal anxiety levels in male rats., Conclusions: Vagal sensory signals from the gastrointestinal tract are critical for baseline and feeding-induced tuning of anxiety via the central amygdala in rats. Our results suggest vagal gut-brain signaling as a target to normalize interoception in anxiety disorders., (Copyright © 2022 Society of Biological Psychiatry. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
25. Oxytocin and cardiometabolic interoception: Knowing oneself affects ingestive and social behaviors.
- Author
-
Smith JA, Eikenberry SA, Scott KA, Baumer-Harrison C, de Lartigue G, de Kloet AD, and Krause EG
- Abstract
Maintaining homeostasis while navigating one's environment involves accurately assessing and interacting with external stimuli while remaining consciously in tune with internal signals such as hunger and thirst. Both atypical social interactions and unhealthy eating patterns emerge as a result of dysregulation in factors that mediate the prioritization and attention to salient stimuli. Oxytocin is an evolutionarily conserved peptide that regulates attention to exteroceptive and interoceptive stimuli in a social environment by functioning in the brain as a modulatory neuropeptide to control social behavior, but also in the periphery as a hormone acting at oxytocin receptors (Oxtr) expressed in the heart, gut, and peripheral ganglia. Specialized sensory afferent nerve endings of Oxtr-expressing nodose ganglia cells transmit cardiometabolic signals via the Vagus nerve to integrative regions in the brain that also express Oxtr(s). These brain regions are influenced by vagal sensory pathways and coordinate with external events such as those demanding attention to social stimuli, thus the sensations related to cardiometabolic function and social interactions are influenced by oxytocin signaling. This review investigates the literature supporting the idea that oxytocin mediates the interoception of cardiovascular and gastrointestinal systems, and that the modulation of this awareness likewise influences social cognition. These concepts are then considered in relation to Autism Spectrum Disorder, exploring how atypical social behavior is comorbid with cardiometabolic dysfunction., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
26. Reappraising the role of the vagus nerve in GLP-1-mediated regulation of eating.
- Author
-
Brierley DI and de Lartigue G
- Subjects
- Humans, Obesity drug therapy, Signal Transduction, Vagus Nerve metabolism, Glucagon-Like Peptide 1, Glucagon-Like Peptide-1 Receptor metabolism
- Abstract
Here, we provide a focused review of the evidence for the roles of the vagus nerve in mediating the regulatory effects of peripherally and centrally produced GLP-1 on eating behaviour and energy balance. We particularly focus on recent studies which have used selective genetic, viral, and transcriptomic approaches to provide important insights into the anatomical and functional organisation of GLP-1-mediated gut-brain signalling pathways. A number of these studies have challenged canonical ideas of how GLP-1 acts in the periphery and the brain to regulate eating behaviour, with important implications for the development of pharmacological treatments for obesity. LINKED ARTICLES: This article is part of a themed issue on GLP1 receptor ligands (BJP 75th Anniversary). To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.4/issuetoc., (© 2021 The Authors. British Journal of Pharmacology published by John Wiley & Sons Ltd on behalf of British Pharmacological Society.)
- Published
- 2022
- Full Text
- View/download PDF
27. Introduction to special issue on feeding peptides.
- Author
-
de Lartigue G, Raybould H, and Dockray G
- Subjects
- Animals, Eating drug effects, Eating physiology, Feeding Behavior, Humans, Neuropeptides physiology, Obesity physiopathology, Peptides pharmacology, Peptides physiology
- Published
- 2022
- Full Text
- View/download PDF
28. Demystifying functional role of cocaine- and amphetamine-related transcript (CART) peptide in control of energy homeostasis: A twenty-five year expedition.
- Author
-
Singh A, de Araujo AM, Krieger JP, Vergara M, Ip CK, and de Lartigue G
- Subjects
- Animals, Energy Metabolism, Homeostasis, Humans, Nerve Tissue Proteins metabolism, Neurons metabolism, Peptides metabolism, Vagus Nerve metabolism
- Abstract
Cocaine- and amphetamine-related transcript (CART) is a neuropeptide first discovered in the striatum of the rat brain. Later, the genetic sequence and function of CART peptide (CARTp) was found to be conserved among multiple mammalian species. Over the 25 years, since its discovery, CART mRNA (Cartpt) expression has been reported widely throughout the central and peripheral nervous systems underscoring its role in diverse physiological functions. Here, we review the localization and function of CARTp as it relates to energy homeostasis. We summarize the expression changes of central and peripheral Cartpt in response to metabolic states and make use of available large data sets to gain additional insights into the anatomy of the Cartpt expressing vagal neurons and their expression patterns in the gut. Furthermore, we provide an overview of the role of CARTp as an anorexigenic signal and its effect on energy expenditure and body weight control with insights from both pharmacological and transgenic animal studies. Subsequently, we discuss the role of CARTp in the pathophysiology of obesity and review important new developments towards identifying a candidate receptor for CARTp signalling. Altogether, the field of CARTp research has made rapid and substantial progress recently, and we review the case for considering CARTp as a potential therapeutic target for stemming the obesity epidemic., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
29. Intact vagal gut-brain signalling prevents hyperphagia and excessive weight gain in response to high-fat high-sugar diet.
- Author
-
McDougle M, Quinn D, Diepenbroek C, Singh A, de la Serre C, and de Lartigue G
- Subjects
- Animals, Brain, Diet, High-Fat adverse effects, Rats, Vagus Nerve, Weight Gain, Hyperphagia, Sugars
- Abstract
Aim: The tools that have been used to assess the function of the vagus nerve lack specificity. This could explain discrepancies about the role of vagal gut-brain signalling in long-term control of energy balance. Here we use a validated approach to selectively ablate sensory vagal neurones that innervate the gut to determine the role of vagal gut-brain signalling in the control of food intake, energy expenditure and glucose homoeostasis in response to different diets., Methods: Rat nodose ganglia were injected bilaterally with either the neurotoxin saporin conjugated to the gastrointestinal hormone cholecystokinin (CCK), or unconjugated saporin as a control. Food intake, body weight, glucose tolerance and energy expenditure were measured in both groups in response to chow or high-fat high-sugar (HFHS) diet. Willingness to work for fat or sugar was assessed by progressive ratio for orally administered solutions, while post-ingestive feedback was tested by measuring food intake after an isocaloric lipid or sucrose pre-load., Results: Vagal deafferentation of the gut increases meal number in lean chow-fed rats. Switching to a HFHS diet exacerbates overeating and body weight gain. The breakpoint for sugar or fat solution did not differ between groups, suggesting that increased palatability may not drive HFHS-induced hyperphagia. Instead, decreased satiation in response to intra-gastric infusion of fat, but not sugar, promotes hyperphagia in CCK-Saporin-treated rats fed with HFHS diet., Conclusions: We conclude that intact sensory vagal neurones prevent hyperphagia and exacerbation of weight gain in response to a HFHS diet by promoting lipid-mediated satiation., (© 2020 The Authors. Acta Physiologica published by John Wiley & Sons Ltd on behalf of Scandinavian Physiological Society.)
- Published
- 2021
- Full Text
- View/download PDF
30. The Medullary Targets of Neurally Conveyed Sensory Information from the Rat Hepatic Portal and Superior Mesenteric Veins.
- Author
-
Garcia-Luna C, Sanchez-Watts G, Arnold M, de Lartigue G, DeWalt N, Langhans W, and Watts AG
- Subjects
- Animals, Male, Neurons, Rats, Solitary Nucleus, Vagus Nerve, Mesenteric Veins, Nodose Ganglion
- Abstract
Vagal and spinal sensory endings in the wall of the hepatic portal and superior mesenteric veins (PMV) provide the brain with chemosensory information important for energy balance and other functions. To determine their medullary neuronal targets, we injected the transsynaptic anterograde viral tracer HSV-1 H129-772 (H129) into the PMV wall or left nodose ganglion (LNG) of male rats, followed by immunohistochemistry (IHC) and high-resolution imaging. We also determined the chemical phenotype of H129-infected neurons, and potential vagal and spinal axon terminal appositions in the dorsal motor nucleus of the vagus (DMX) and the nucleus of the solitary tract (NTS). PMV wall injections generated H129-infected neurons in both nodose ganglia and in thoracic dorsal root ganglia (DRGs). In the medulla, cholinergic preganglionic parasympathetic neurons in the DMX were virtually the only targets of chemosensory information from the PMV wall. H129-infected terminal appositions were identified on H129-infected somata and dendrites in the DMX, and on H129-infected DMX dendrites that extend into the NTS. Sensory transmission via vagal and possibly spinal routes from the PMV wall therefore reaches DMX neurons via axo-somatic appositions in the DMX and axo-dendritic appositions in the NTS. However, the dearth of H129-infected NTS neurons indicates that sensory information from the PMV wall terminates on DMX neurons without engaging NTS neurons. These previously underappreciated direct sensory routes into the DMX enable a vago-vagal and possibly spino-vagal reflexes that can directly influence visceral function., (Copyright © 2021 Garcia-Luna et al.)
- Published
- 2021
- Full Text
- View/download PDF
31. Central and peripheral GLP-1 systems independently suppress eating.
- Author
-
Brierley DI, Holt MK, Singh A, de Araujo A, McDougle M, Vergara M, Afaghani MH, Lee SJ, Scott K, Maske C, Langhans W, Krause E, de Kloet A, Gribble FM, Reimann F, Rinaman L, de Lartigue G, and Trapp S
- Subjects
- Animals, Eating, Female, Gastrointestinal Tract innervation, Gastrointestinal Tract physiology, Glucagon-Like Peptide-1 Receptor agonists, Glucagon-Like Peptides pharmacology, Male, Mice, Mice, Inbred C57BL, Neurons metabolism, Proglucagon metabolism, Receptors, Oxytocin metabolism, Vagus Nerve physiology, Central Nervous System physiology, Glucagon-Like Peptide 1 physiology, Peripheral Nervous System physiology, Satiety Response physiology
- Abstract
The anorexigenic peptide glucagon-like peptide-1 (GLP-1) is secreted from gut enteroendocrine cells and brain preproglucagon (PPG) neurons, which, respectively, define the peripheral and central GLP-1 systems. PPG neurons in the nucleus tractus solitarii (NTS) are widely assumed to link the peripheral and central GLP-1 systems in a unified gut-brain satiation circuit. However, direct evidence for this hypothesis is lacking, and the necessary circuitry remains to be demonstrated. Here we show that PPG
NTS neurons encode satiation in mice, consistent with vagal signalling of gastrointestinal distension. However, PPGNTS neurons predominantly receive vagal input from oxytocin-receptor-expressing vagal neurons, rather than those expressing GLP-1 receptors. PPGNTS neurons are not necessary for eating suppression by GLP-1 receptor agonists, and concurrent PPGNTS neuron activation suppresses eating more potently than semaglutide alone. We conclude that central and peripheral GLP-1 systems suppress eating via independent gut-brain circuits, providing a rationale for pharmacological activation of PPGNTS neurons in combination with GLP-1 receptor agonists as an obesity treatment strategy.- Published
- 2021
- Full Text
- View/download PDF
32. tVNS Increases Liking of Orally Sampled Low-Fat Foods: A Pilot Study.
- Author
-
Öztürk L, Büning PE, Frangos E, de Lartigue G, and Veldhuizen MG
- Abstract
Recently a role for the vagus nerve in conditioning food preferences was established in rodents. In a prospective controlled clinical trial in humans, invasive vagus nerve stimulation shifted food choice toward lower fat content. Here we explored whether hedonic aspects of an orally sampled food stimulus can be modulated by non-invasive transcutaneous vagus nerve stimulation (tVNS) in humans. In healthy participants ( n = 10, five women, 20-32 years old, no obesity) we tested liking and wanting ratings of food samples with varying fat or sugar content with or without tVNS in a sham-controlled within-participants design. To determine effects of tVNS on food intake, we also measured voluntary consumption of milkshake. Spontaneous eye blink rate was measured as a proxy for dopamine tone. Liking of low-fat, but not high-fat puddings, was higher for tVNS relative to sham stimulation. Other outcomes showed no differences. These findings support a role for the vagus nerve promoting post-ingestive reward signals. Our results suggest that tVNS may be used to increase liking of low-calorie foods, which may support healthier food choices., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2020 Öztürk, Büning, Frangos, de Lartigue and Veldhuizen.)
- Published
- 2020
- Full Text
- View/download PDF
33. Ghrelin Signaling Affects Feeding Behavior, Metabolism, and Memory through the Vagus Nerve.
- Author
-
Davis EA, Wald HS, Suarez AN, Zubcevic J, Liu CM, Cortella AM, Kamitakahara AK, Polson JW, Arnold M, Grill HJ, de Lartigue G, and Kanoski SE
- Subjects
- Afferent Pathways physiology, Animals, Body Weight physiology, Brain-Derived Neurotrophic Factor metabolism, Feeding Behavior physiology, Gastric Emptying physiology, Gene Knockdown Techniques, Glucose metabolism, Hunger physiology, Male, Memory, Episodic, Mice, Models, Animal, Neurons metabolism, Nodose Ganglion cytology, Nodose Ganglion surgery, Rats, Rats, Transgenic, Receptors, Ghrelin genetics, Vagotomy, Ghrelin metabolism, Hippocampus physiology, Nodose Ganglion metabolism, Receptors, Ghrelin metabolism
- Abstract
Vagal afferent neuron (VAN) signaling sends information from the gut to the brain and is fundamental in the control of feeding behavior and metabolism [1]. Recent findings reveal that VAN signaling also plays a critical role in cognitive processes, including affective motivational behaviors and hippocampus (HPC)-dependent memory [2-5]. VANs, located in nodose ganglia, express receptors for various gut-derived peptide signals; however, the function of these receptors with regard to feeding behavior, metabolism, and memory control is poorly understood. We hypothesized that VAN-mediated processes are influenced by ghrelin, a stomach-derived orexigenic hormone, via communication to its receptor (GHSR) expressed on gut-innervating VANs. To examine this hypothesis, rats received nodose ganglia injections of an adeno-associated virus (AAV) expressing short hairpin RNAs targeting GHSR (or a control AAV) for RNAi-mediated VAN-specific GHSR knockdown. Results reveal that VAN GHSR knockdown induced various feeding and metabolic disturbances, including increased meal frequency, impaired glucose tolerance, delayed gastric emptying, and increased body weight compared to controls. Additionally, VAN-specific GHSR knockdown impaired HPC-dependent contextual episodic memory and reduced HPC brain-derived neurotrophic factor expression, but did not affect anxiety-like behavior or general activity levels. A functional role for endogenous VAN GHSR signaling was further confirmed by results revealing that VAN signaling is required for the hyperphagic effects of ghrelin administered at dark onset, and that gut-restricted ghrelin-induced increases in VAN firing rate require intact VAN GHSR expression. Collective results reveal that VAN GHSR signaling is required for both normal feeding and metabolic function as well as HPC-dependent memory., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
34. Non-canonical cholinergic anti-inflammatory pathway in IBD.
- Author
-
de Araujo A and de Lartigue G
- Subjects
- Brain, Humans, Inflammation, Liver, Neuroimmunomodulation, T-Lymphocytes, Regulatory, Gastrointestinal Microbiome, Inflammatory Bowel Diseases drug therapy
- Published
- 2020
- Full Text
- View/download PDF
35. Dissecting the Role of Subtypes of Gastrointestinal Vagal Afferents.
- Author
-
Wang YB, de Lartigue G, and Page AJ
- Abstract
Gastrointestinal (GI) vagal afferents convey sensory signals from the GI tract to the brain. Numerous subtypes of GI vagal afferent have been identified but their individual roles in gut function and feeding regulation are unclear. In the past decade, technical approaches to selectively target vagal afferent subtypes and to assess their function has significantly progressed. This review examines the classification of GI vagal afferent subtypes and discusses the current available techniques to study vagal afferents. Investigating the distribution of GI vagal afferent subtypes and understanding how to access and modulate individual populations are essential to dissect their fundamental roles in the gut-brain axis., (Copyright © 2020 Wang, de Lartigue and Page.)
- Published
- 2020
- Full Text
- View/download PDF
36. Gut microbiota sPARk vagus nerve excitation.
- Author
-
Singh A, de la Serre C, and de Lartigue G
- Subjects
- Bacteria, Neurons, Nodose Ganglion, Peptide Hydrolases, Vagus Nerve, Gastrointestinal Microbiome
- Published
- 2020
- Full Text
- View/download PDF
37. Blunted Vagal Cocaine- and Amphetamine-Regulated Transcript Promotes Hyperphagia and Weight Gain.
- Author
-
Lee SJ, Krieger JP, Vergara M, Quinn D, McDougle M, de Araujo A, Darling R, Zollinger B, Anderson S, Pan A, Simonnet EJ, Pignalosa A, Arnold M, Singh A, Langhans W, Raybould HE, and de Lartigue G
- Subjects
- Animals, Humans, Male, Rats, Hyperphagia genetics, Nerve Tissue Proteins metabolism, Vagus Nerve drug effects, Weight Gain genetics
- Abstract
The vagus nerve conveys gastrointestinal cues to the brain to control eating behavior. In obesity, vagally mediated gut-brain signaling is disrupted. Here, we show that the cocaine- and amphetamine-regulated transcript (CART) is a neuropeptide synthesized proportional to the food consumed in vagal afferent neurons (VANs) of chow-fed rats. CART injection into the nucleus tractus solitarii (NTS), the site of vagal afferent central termination, reduces food intake. Conversely, blocking endogenous CART action in the NTS increases food intake in chow-fed rats, and this requires intact VANs. Viral-mediated Cartpt knockdown in VANs increases weight gain and daily food intake via larger meals and faster ingestion rate. In obese rats fed a high-fat, high-sugar diet, meal-induced CART synthesis in VANs is blunted and CART antibody fails to increase food intake. However, CART injection into the NTS retains its anorexigenic effect in obese rats. Restoring disrupted VAN CART signaling in obesity could be a promising therapeutic approach., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
38. Vagus nerve regulates the phagocytic and secretory activity of resident macrophages in the liver.
- Author
-
Fonseca RC, Bassi GS, Brito CC, Rosa LB, David BA, Araújo AM, Nóbrega N, Diniz AB, Jesus ICG, Barcelos LS, Fontes MAP, Bonaventura D, Kanashiro A, Cunha TM, Guatimosim S, Cardoso VN, Fernandes SOA, Menezes GB, de Lartigue G, and Oliveira AG
- Subjects
- Animals, Cytokines, Female, Gastrointestinal Tract, Liver pathology, Macrophages immunology, Mice, Mice, Inbred C57BL, Phagocytes metabolism, Sepsis immunology, Vagus Nerve pathology, Vagus Nerve Stimulation methods, Liver immunology, Phagocytosis physiology, Vagus Nerve physiology
- Abstract
The gastrointestinal (GI) tract harbors commensal microorganisms as well as invasive bacteria, toxins and other pathogens and, therefore, plays a pivotal barrier and immunological role against pathogenic agents. The vagus nerve is an important regulator of the GI tract-associated immune system, having profound effects on inflammatory responses. Among GI tract organs, the liver is a key site of immune surveillance, as it has a large population of resident macrophages and receives the blood drained from the guts through the hepatic portal circulation. Although it is widely accepted that the hepatic tissue is a major target for vagus nerve fibers, the role of this neural circuit in liver immune functions is still poorly understood. Herein we used in vivo imaging techniques, including confocal microscopy and scintigraphy, to show that vagus nerve stimulation increases the phagocytosis activity by resident macrophages in the liver, even on the absence of an immune challenge. The activation of this neural circuit in a non-lethal model of sepsis optimized the removal of bacteria in the liver and resulted in the production of anti-inflammatory and pro-regenerative cytokines. Our findings provide new insights into the neural regulation of the immune system in the liver., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
39. Deletion of leptin receptors in vagal afferent neurons disrupts estrogen signaling, body weight, food intake and hormonal controls of feeding in female mice.
- Author
-
Huang KP, Ronveaux CC, de Lartigue G, Geary N, Asarian L, and Raybould HE
- Subjects
- Animals, Arcuate Nucleus of Hypothalamus metabolism, Body Weight genetics, Cholecystokinin pharmacology, Diet, High-Fat, Eating drug effects, Energy Metabolism, Estradiol metabolism, Estrogen Receptor alpha metabolism, Estrogens metabolism, Feeding Behavior drug effects, Female, Ghrelin pharmacology, Mice, Obesity metabolism, Satiation, Vagus Nerve cytology, Weight Gain genetics, Eating genetics, Feeding Behavior physiology, Neurons, Afferent metabolism, Obesity genetics, Receptors, Leptin genetics
- Abstract
Deletion of the leptin receptor from vagal afferent neurons (VAN) using a conditional deletion (Nav1.8/LepR
fl/fl ) results in an obese phenotype with increased food intake and lack of exogenous cholecystokinin (CCK)-induced satiation in male mice. Female mice are partially protected from weight gain and increased food intake in response to ingestion of high-fat (HF) diets. However, whether the lack of leptin signaling in VAN leads to an obese phenotype or disruption of hypothalamic-pituitary-gonadal axis function in female mice is unclear. Here, we tested the hypothesis that leptin signaling in VAN is essential to maintain estrogen signaling and control of food intake, energy expenditure, and adiposity in female mice. Female Nav1.8/LepRfl/fl mice gained more weight, had increased gonadal fat mass, increased meal number in the dark phase, and increased total food intake compared with wild-type controls. Resting energy expenditure was unaffected. The decrease in food intake produced by intraperitoneal injection of CCK (3 μg/kg body wt) was attenuated in female Nav1.8/LepRfl/fl mice compared with wild-type controls. Intraperitoneal injection of ghrelin (100 μg/kg body wt) increased food intake in Nav1.8/LepRfl/fl mice but not in wild-type controls. Ovarian steroidogenesis was suppressed, resulting in decreased plasma estradiol, which was accompanied by decreased expression of estrogen receptor-1 (Esr1) in VAN but not in the hypothalamic arcuate nucleus. These data suggest that the absence of leptin signaling in VAN is accompanied by disruption of estrogen signaling in female mice, leading to an obese phenotype possibly via altered control of feeding behavior.- Published
- 2019
- Full Text
- View/download PDF
40. Dorsal striatum dopamine oscillations: Setting the pace of food anticipatory activity.
- Author
-
de Lartigue G and McDougle M
- Subjects
- Animals, Suprachiasmatic Nucleus physiology, Appetite physiology, Circadian Rhythm physiology, Corpus Striatum physiology, Dopamine metabolism, Feeding Behavior physiology
- Abstract
Predicting the uncertainties of the ever-changing environment provides a competitive advantage for animals. The need to anticipate food sources has provided a strong evolutionary drive for synchronizing behavioural and internal processes with daily circadian cycles. When food is restricted to a few hours per day, rodents exhibit increased wakefulness and foraging behaviour preceding the arrival of food. Interestingly, while the master clock located in the suprachiasmatic nucleus entrains daily rhythms to the light cycle, it is not necessary for this food anticipatory activity. This suggests the existence of a food-entrained oscillator located elsewhere. Based on the role of nigrostriatal dopamine in reward processing, motor function, working memory and internal timekeeping, we propose a working model by which food-entrained dopamine oscillations in the dorsal striatum can enable animals maintained on a restricted feeding schedule to anticipate food arrival. Finally, we summarize how metabolic signals in the gut are conveyed to the nigrostriatal pathway to suggest possible insight into potential input mechanisms for food anticipatory activity., (© 2018 Scandinavian Physiological Society. Published by John Wiley & Sons Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
41. A Neural Circuit for Gut-Induced Reward.
- Author
-
Han W, Tellez LA, Perkins MH, Perez IO, Qu T, Ferreira J, Ferreira TL, Quinn D, Liu ZW, Gao XB, Kaelberer MM, Bohórquez DV, Shammah-Lagnado SJ, de Lartigue G, and de Araujo IE
- Subjects
- Afferent Pathways metabolism, Afferent Pathways physiology, Animals, Dopamine metabolism, Dopaminergic Neurons physiology, Glutamic Acid metabolism, Intestines innervation, Male, Mice, Mice, Inbred C57BL, Optogenetics, Intestines physiology, Reward, Substantia Nigra physiology, Vagus Nerve physiology
- Abstract
The gut is now recognized as a major regulator of motivational and emotional states. However, the relevant gut-brain neuronal circuitry remains unknown. We show that optical activation of gut-innervating vagal sensory neurons recapitulates the hallmark effects of stimulating brain reward neurons. Specifically, right, but not left, vagal sensory ganglion activation sustained self-stimulation behavior, conditioned both flavor and place preferences, and induced dopamine release from Substantia nigra. Cell-specific transneuronal tracing revealed asymmetric ascending pathways of vagal origin throughout the CNS. In particular, transneuronal labeling identified the glutamatergic neurons of the dorsolateral parabrachial region as the obligatory relay linking the right vagal sensory ganglion to dopamine cells in Substantia nigra. Consistently, optical activation of parabrachio-nigral projections replicated the rewarding effects of right vagus excitation. Our findings establish the vagal gut-to-brain axis as an integral component of the neuronal reward pathway. They also suggest novel vagal stimulation approaches to affective disorders., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
42. Mechanisms of vagal plasticity influencing feeding behavior.
- Author
-
de Lartigue G and Xu C
- Subjects
- Animals, Humans, Neuropeptides metabolism, Feeding Behavior physiology, Neuronal Plasticity physiology, Vagus Nerve cytology, Vagus Nerve physiology
- Abstract
Sensory neurons of the vagus nerve receive many different peripheral signals that can change rapidly and frequently throughout the day. The ability of these neurons to convey the vast array of nuanced information to the brain requires neuronal adaptability. In this review we discuss evidence for neural plasticity in vagal afferent neurons as a mechanism for conveying nuanced information to the brain important for the control of feeding behavior. We provide evidence that synaptic plasticity, changes in membrane conductance, and neuropeptide specification are mechanisms that allow flexibility in response to metabolic cues that can be disrupted by chronic intake of energy dense diets., (Copyright © 2018. Published by Elsevier B.V.)
- Published
- 2018
- Full Text
- View/download PDF
43. New horizons for future research - Critical issues to consider for maximizing research excellence and impact.
- Author
-
Langhans W, Adan R, Arnold M, Banks WA, Card JP, Dailey MJ, Daniels D, de Kloet AD, de Lartigue G, Dickson S, Fedele S, Grill HJ, Jansson JO, Kaufman S, Kolar G, Krause E, Lee SJ, Le Foll C, Levin BE, Lutz TA, Mansouri A, Moran TH, Pacheco-López G, Ramachandran D, Raybould H, Rinaman L, Samson WK, Sanchez-Watts G, Seeley RJ, Skibicka KP, Small D, Spector AC, Tamashiro KL, Templeton B, Trapp S, Tso P, Watts AG, Weissfeld N, Williams D, Wolfrum C, Yosten G, and Woods SC
- Subjects
- Biomedical Research economics, Biomedical Research education, Biomedical Research standards, Education, Professional standards, Financing, Organized standards, Peer Review, Research standards, Quality Control, Research Design standards, Biomedical Research trends
- Published
- 2018
- Full Text
- View/download PDF
44. Gut vagal sensory signaling regulates hippocampus function through multi-order pathways.
- Author
-
Suarez AN, Hsu TM, Liu CM, Noble EE, Cortella AM, Nakamoto EM, Hahn JD, de Lartigue G, and Kanoski SE
- Subjects
- Animals, Cerebral Cortex physiology, Male, Memory physiology, Rats, Sprague-Dawley, Synapses physiology, Telencephalon physiology, Gastrointestinal Tract innervation, Hippocampus physiology, Neural Pathways physiology, Sensory Receptor Cells physiology, Vagus Nerve physiology
- Abstract
The vagus nerve is the primary means of neural communication between the gastrointestinal (GI) tract and the brain. Vagally mediated GI signals activate the hippocampus (HPC), a brain region classically linked with memory function. However, the endogenous relevance of GI-derived vagal HPC communication is unknown. Here we utilize a saporin (SAP)-based lesioning procedure to reveal that selective GI vagal sensory/afferent ablation in rats impairs HPC-dependent episodic and spatial memory, effects associated with reduced HPC neurotrophic and neurogenesis markers. To determine the neural pathways connecting the gut to the HPC, we utilize monosynaptic and multisynaptic virus-based tracing methods to identify the medial septum as a relay connecting the medial nucleus tractus solitarius (where GI vagal afferents synapse) to dorsal HPC glutamatergic neurons. We conclude that endogenous GI-derived vagal sensory signaling promotes HPC-dependent memory function via a multi-order brainstem-septal pathway, thereby identifying a previously unknown role for the gut-brain axis in memory control.
- Published
- 2018
- Full Text
- View/download PDF
45. Validation and characterization of a novel method for selective vagal deafferentation of the gut.
- Author
-
Diepenbroek C, Quinn D, Stephens R, Zollinger B, Anderson S, Pan A, and de Lartigue G
- Subjects
- Afferent Pathways physiology, Animals, Gastrointestinal Tract physiology, Male, Neurotoxins administration & dosage, Neurotoxins pharmacology, Rats, Rats, Wistar, Saporins, Treatment Outcome, Vagus Nerve physiology, Afferent Pathways drug effects, Autonomic Denervation methods, Gastrointestinal Tract drug effects, Nerve Block methods, Ribosome Inactivating Proteins, Type 1 administration & dosage, Vagus Nerve drug effects
- Abstract
There is a lack of tools that selectively target vagal afferent neurons (VAN) innervating the gut. We use saporin (SAP), a potent neurotoxin, conjugated to the gastronintestinal (GI) hormone cholecystokinin (CCK-SAP) injected into the nodose ganglia (NG) of male Wistar rats to specifically ablate GI-VAN. We report that CCK-SAP ablates a subpopulation of VAN in culture. In vivo, CCK-SAP injection into the NG reduces VAN innervating the mucosal and muscular layers of the stomach and small intestine but not the colon, while leaving vagal efferent neurons intact. CCK-SAP abolishes feeding-induced c-Fos in the NTS, as well as satiation by CCK or glucagon like peptide-1 (GLP-1). CCK-SAP in the NG of mice also abolishes CCK-induced satiation. Therefore, we provide multiple lines of evidence that injection of CCK-SAP in NG is a novel selective vagal deafferentation technique of the upper GI tract that works in multiple vertebrate models. This method provides improved tissue specificity and superior separation of afferent and efferent signaling compared with vagotomy, capsaicin, and subdiaphragmatic deafferentation. NEW & NOTEWORTHY We develop a new method that allows targeted lesioning of vagal afferent neurons that innervate the upper GI tract while sparing vagal efferent neurons. This reliable approach provides superior tissue specificity and selectivity for vagal afferent over efferent targeting than traditional approaches. It can be used to address questions about the role of gut to brain signaling in physiological and pathophysiological conditions., (Copyright © 2017 the American Physiological Society.)
- Published
- 2017
- Full Text
- View/download PDF
46. XIVth Little Brain Big Brain: next-generation enteric neuroscience.
- Author
-
Beyder A, de Lartigue G, Ghia JE, and Hoffman JM
- Subjects
- Brain physiopathology, Congresses as Topic, Enteric Nervous System embryology, Enteric Nervous System microbiology, Gastrointestinal Microbiome, Gastrointestinal Motility, Humans, Intestinal Diseases etiology, Intestinal Diseases physiopathology, Metabolic Diseases etiology, Metabolic Diseases physiopathology, Neurosciences, Enteric Nervous System physiopathology
- Abstract
Little Brain Big Brain has been a biannual meeting organized and attended exclusively by young investigators in neurogastroenterology since 1989. The XIVth meeting featured cutting-edge work advancing several novel hypotheses in the main themes of motility, inflammation and metabolism.
- Published
- 2017
- Full Text
- View/download PDF
47. Novel developments in vagal afferent nutrient sensing and its role in energy homeostasis.
- Author
-
de Lartigue G and Diepenbroek C
- Subjects
- Animals, Body Weight physiology, Circadian Rhythm physiology, Eating physiology, Ghrelin metabolism, Glucagon-Like Peptide 1 metabolism, Homeostasis physiology, Humans, Reward, Signal Transduction physiology, Energy Metabolism physiology, Neurons, Afferent metabolism, Vagus Nerve physiology
- Abstract
Vagal afferent neurons (VANs) play an important role in the control of food intake by signaling nutrient type and quantity to the brain. Recent findings are broadening our view of how VANs impact not only food intake but also energy homeostasis. This review focuses exclusively on studies of the vagus nerve from the past 2 years that highlight major new advancements in the field. We firstly discuss evidence that VANs can directly sense nutrients, and we consider new insights into mechanisms affecting sensing of gastric distension and signaling by gastrointestinal hormones ghrelin and GLP1. We discuss evidence that disrupting vagal afferent signaling increases long-term control of food intake and body weight management, and the importance of this gut-brain pathway in mediating beneficial effects of bariatric surgery. We conclude by highlighting novel roles for vagal afferent neurons in circadian rhythm, thermogenesis, and reward that may provide insight into mechanisms by which VAN nutrient sensing controls long-term control of energy homeostasis., (Copyright © 2016. Published by Elsevier Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
48. Role of the vagus nerve in the development and treatment of diet-induced obesity.
- Author
-
de Lartigue G
- Subjects
- Animals, Body Weight physiology, Diet, High-Fat, Humans, Hyperphagia physiopathology, Neurons, Afferent physiology, Obesity physiopathology, Vagus Nerve physiology
- Abstract
This review highlights evidence for a role of the vagus nerve in the development of obesity and how targeting the vagus nerve with neuromodulation or pharmacology can be used as a therapeutic treatment of obesity. The vagus nerve innervating the gut plays an important role in controlling metabolism. It communicates peripheral information about the volume and type of nutrients between the gut and the brain. Depending on the nutritional status, vagal afferent neurons express two different neurochemical phenotypes that can inhibit or stimulate food intake. Chronic ingestion of calorie-rich diets reduces sensitivity of vagal afferent neurons to peripheral signals and their constitutive expression of orexigenic receptors and neuropeptides. This disruption of vagal afferent signalling is sufficient to drive hyperphagia and obesity. Furthermore neuromodulation of the vagus nerve can be used in the treatment of obesity. Although the mechanisms are poorly understood, vagal nerve stimulation prevents weight gain in response to a high-fat diet. In small clinical studies, in patients with depression or epilepsy, vagal nerve stimulation has been demonstrated to promote weight loss. Vagal blockade, which inhibits the vagus nerve, results in significant weight loss. Vagal blockade is proposed to inhibit aberrant orexigenic signals arising in obesity as a putative mechanism of vagal blockade-induced weight loss. Approaches and molecular targets to develop future pharmacotherapy targeted to the vagus nerve for the treatment of obesity are proposed. In conclusion there is strong evidence that the vagus nerve is involved in the development of obesity and it is proving to be an attractive target for the treatment of obesity., (© 2016 The Authors. The Journal of Physiology © 2016 The Physiological Society.)
- Published
- 2016
- Full Text
- View/download PDF
49. Chronic exposure to low dose bacterial lipopolysaccharide inhibits leptin signaling in vagal afferent neurons.
- Author
-
de La Serre CB, de Lartigue G, and Raybould HE
- Subjects
- Animals, Blotting, Western, Hyperphagia physiopathology, Immunohistochemistry, Male, Peroxidase metabolism, Rats, Wistar, Satiation physiology, Sincalide administration & dosage, Sincalide metabolism, Weight Gain physiology, Eating physiology, Leptin metabolism, Lipopolysaccharides toxicity, Neurons, Afferent physiology, Nodose Ganglion physiopathology
- Abstract
Bacterially derived factors are implicated in the causation and persistence of obesity. Ingestion of a high fat diet in rodents and obesity in human subjects is associated with chronic elevation of low plasma levels of lipopolysaccharide (LPS), a breakdown product of Gram-negative bacteria. The terminals of vagal afferent neurons are positioned within the gut mucosa to convey information from the gut to the brain to regulate food intake and are responsive to LPS. We hypothesized that chronic elevation of LPS could alter vagal afferent signaling. We surgically implanted osmotic mini-pumps that delivered a constant, low-dose of LPS into the intraperitoneal cavity of rats (12.5 μg/kg/hr for 6 weeks). LPS-treated rats developed hyperphagia and showed marked changes in vagal afferent neuron function. Chronic LPS treatment reduced vagal afferent leptin signaling, characterized by a decrease in leptin-induced STAT3 phosphorylation. In addition, LPS treatment decreased cholecystokinin-induced satiety. There was no alteration in leptin signaling in the hypothalamus. These findings offer a mechanism by which a change in gut microflora can promote hyperphagia, possibly leading to obesity., (Copyright © 2014. Published by Elsevier Inc.)
- Published
- 2015
- Full Text
- View/download PDF
50. Vagal plasticity the key to obesity.
- Author
-
de Lartigue G, Ronveaux CC, and Raybould HE
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.