1
|
Ramasamy I. Physiological Appetite Regulation and Bariatric Surgery. J Clin Med 2024; 13:1347. [PMID: 38546831 PMCID: PMC10932430 DOI: 10.3390/jcm13051347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 02/22/2024] [Accepted: 02/24/2024] [Indexed: 04/10/2024] Open
Abstract
Obesity remains a common metabolic disorder and a threat to health as it is associated with numerous complications. Lifestyle modifications and caloric restriction can achieve limited weight loss. Bariatric surgery is an effective way of achieving substantial weight loss as well as glycemic control secondary to weight-related type 2 diabetes mellitus. It has been suggested that an anorexigenic gut hormone response following bariatric surgery contributes to weight loss. Understanding the changes in gut hormones and their contribution to weight loss physiology can lead to new therapeutic treatments for weight loss. Two distinct types of neurons in the arcuate hypothalamic nuclei control food intake: proopiomelanocortin neurons activated by the anorexigenic (satiety) hormones and neurons activated by the orexigenic peptides that release neuropeptide Y and agouti-related peptide (hunger centre). The arcuate nucleus of the hypothalamus integrates hormonal inputs from the gut and adipose tissue (the anorexigenic hormones cholecystokinin, polypeptide YY, glucagon-like peptide-1, oxyntomodulin, leptin, and others) and orexigeneic peptides (ghrelin). Replicating the endocrine response to bariatric surgery through pharmacological mimicry holds promise for medical treatment. Obesity has genetic and environmental factors. New advances in genetic testing have identified both monogenic and polygenic obesity-related genes. Understanding the function of genes contributing to obesity will increase insights into the biology of obesity. This review includes the physiology of appetite control, the influence of genetics on obesity, and the changes that occur following bariatric surgery. This has the potential to lead to the development of more subtle, individualised, treatments for obesity.
Collapse
Affiliation(s)
- Indra Ramasamy
- Department of Blood Sciences, Conquest Hospital, Hastings TN37 7RD, UK
| |
Collapse
|
2
|
Fan C, Xu J, Tong H, Fang Y, Chen Y, Lin Y, Chen R, Chen F, Wu G. Gut-brain communication mediates the impact of dietary lipids on cognitive capacity. Food Funct 2024; 15:1803-1824. [PMID: 38314832 DOI: 10.1039/d3fo05288e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Cognitive impairment, as a prevalent symptom of nervous system disorders, poses one of the most challenging aspects in the management of brain diseases. Lipids present in the cell membranes of all neurons within the brain and dietary lipids can regulate the cognition and memory function. In recent years, the advancements in gut microbiome research have enabled the exploration of dietary lipids targeting the gut-brain axis as a strategy for regulating cognition. This present review provides an in-depth overview of how lipids modulate cognition via the gut-brain axis depending on metabolic, immune, neural and endocrine pathways. It also comprehensively analyzes the effects of diverse lipids on the gut microbiota and intestinal barrier function, thereby affecting the central nervous system and cognitive capacity. Moreover, comparative analysis of the positive and negative effects is presented between beneficial and detrimental lipids. The former encompass monounsaturated fatty acids, short-chain fatty acids, omega-3 polyunsaturated fatty acids, phospholipids, phytosterols, fungal sterols and bioactive lipid-soluble vitamins, as well as lipid-derived gut metabolites, whereas the latter (detrimental lipids) include medium- or long-chain fatty acids, excessive proportions of n-6 polyunsaturated fatty acids, industrial trans fatty acids, and zoosterols. To sum up, the focus of this review is on how gut-brain communication mediates the impact of dietary lipids on cognitive capacity, providing a novel theoretical foundation for promoting brain cognitive health and scientific lipid consumption patterns.
Collapse
Affiliation(s)
- Chenhan Fan
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Jingxuan Xu
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Haoxiang Tong
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Yucheng Fang
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Yiming Chen
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Yangzhuo Lin
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China
| | - Rui Chen
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China
| | - Fuhao Chen
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China
| | - Guoqing Wu
- School of Public Health, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China.
| |
Collapse
|
3
|
Eleftheriadis PE, Pothakos K, Sharples SA, Apostolou PE, Mina M, Tetringa E, Tsape E, Miles GB, Zagoraiou L. Peptidergic modulation of motor neuron output via CART signaling at C bouton synapses. Proc Natl Acad Sci U S A 2023; 120:e2300348120. [PMID: 37733738 PMCID: PMC10523464 DOI: 10.1073/pnas.2300348120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 07/17/2023] [Indexed: 09/23/2023] Open
Abstract
The intensity of muscle contraction, and therefore movement vigor, needs to be adaptable to enable complex motor behaviors. This can be achieved by adjusting the properties of motor neurons, which form the final common pathway for all motor output from the central nervous system. Here, we identify roles for a neuropeptide, cocaine- and amphetamine-regulated transcript (CART), in the control of movement vigor. We reveal distinct but parallel mechanisms by which CART and acetylcholine, both released at C bouton synapses on motor neurons, selectively amplify the output of subtypes of motor neurons that are recruited during intense movement. We find that mice with broad genetic deletion of CART or selective elimination of acetylcholine from C boutons exhibit deficits in behavioral tasks that require higher levels of motor output. Overall, these data uncover spinal modulatory mechanisms that control movement vigor to support movements that require a high degree of muscle force.
Collapse
Affiliation(s)
| | - Konstantinos Pothakos
- Center of Basic Research, Biomedical Research Foundation Academy of Athens, Athens11527, Greece
| | - Simon A. Sharples
- School of Psychology and Neuroscience, University of St. Andrews, St. AndrewsKY16 9JP, United Kingdom
| | - Panagiota E. Apostolou
- Center of Basic Research, Biomedical Research Foundation Academy of Athens, Athens11527, Greece
| | - Maria Mina
- Center of Basic Research, Biomedical Research Foundation Academy of Athens, Athens11527, Greece
| | - Efstathia Tetringa
- Center of Basic Research, Biomedical Research Foundation Academy of Athens, Athens11527, Greece
| | - Eirini Tsape
- Center of Basic Research, Biomedical Research Foundation Academy of Athens, Athens11527, Greece
| | - Gareth B. Miles
- School of Psychology and Neuroscience, University of St. Andrews, St. AndrewsKY16 9JP, United Kingdom
| | - Laskaro Zagoraiou
- Center of Basic Research, Biomedical Research Foundation Academy of Athens, Athens11527, Greece
| |
Collapse
|
4
|
Wierup N, Abels M, Shcherbina L, Lindqvist A. The role of CART in islet biology. Peptides 2022; 149:170708. [PMID: 34896575 DOI: 10.1016/j.peptides.2021.170708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/03/2021] [Accepted: 12/03/2021] [Indexed: 10/19/2022]
Abstract
Cocaine- and amphetamine-regulated transcript (CART) is mostly known for its appetite regulating effects in the central nervous system. However, CART is also highly expressed in the peripheral nervous system as well as in certain endocrine cells. Our group has dedicated more than 20 years to understand the role of CART in the pancreatic islets and in this review we summarize what is known to date about CART expression and function in the islets. CART is expressed in both islet cells and nerve fibers innervating the islets. Large species differences are at hand and CART expression is highly dynamic and increased during development, as well as in Type 2 Diabetes and certain endocrine tumors. In the human islets CART is expressed in alpha cells and beta cells and the expression is increased in T2D patients. CART increases insulin secretion, reduces glucagon secretion, and protects against beta cell death by reducing apoptosis and increasing proliferation. It is still not fully understood how CART mediates its effects or which receptors that are involved. Nevertheless, CART is endowed with several properties that are beneficial in a T2D perspective. Many of the described effects of CART resemble those of GLP-1, and interestingly CART has been found to potentiate some of the effects of GLP-1, paving the way for CART-based treatments in combination with GLP-1-based drugs.
Collapse
Affiliation(s)
- Nils Wierup
- Lund University Diabetes Centre, Malmö, Sweden.
| | - Mia Abels
- Lund University Diabetes Centre, Malmö, Sweden
| | | | | |
Collapse
|
5
|
Singh A, de Araujo AM, Krieger JP, Vergara M, Ip CK, de Lartigue G. Demystifying functional role of cocaine- and amphetamine-related transcript (CART) peptide in control of energy homeostasis: A twenty-five year expedition. Peptides 2021; 140:170534. [PMID: 33757831 PMCID: PMC8369463 DOI: 10.1016/j.peptides.2021.170534] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 02/28/2021] [Accepted: 03/16/2021] [Indexed: 12/17/2022]
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.
Collapse
Affiliation(s)
- Arashdeep Singh
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA; Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL, USA
| | - Alan Moreira de Araujo
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA; Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL, USA
| | - Jean-Philippe Krieger
- Department of Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Macarena Vergara
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA; Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL, USA
| | - Chi Kin Ip
- Neuroscience Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia; Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Guillaume de Lartigue
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA; Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
6
|
Cawthon CR, de La Serre CB. The critical role of CCK in the regulation of food intake and diet-induced obesity. Peptides 2021; 138:170492. [PMID: 33422646 DOI: 10.1016/j.peptides.2020.170492] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 12/16/2020] [Accepted: 12/23/2020] [Indexed: 12/17/2022]
Abstract
In 1973, Gibbs, Young, and Smith showed that exogenous cholecystokinin (CCK) administration reduces food intake in rats. This initial report has led to thousands of studies investigating the physiological role of CCK in regulating feeding behavior. CCK is released from enteroendocrine I cells present along the gastrointestinal (GI) tract. CCK binding to its receptor CCK1R leads to vagal afferent activation providing post-ingestive feedback to the hindbrain. Vagal afferent neurons' (VAN) sensitivity to CCK is modulated by energy status while CCK signaling regulates gene expression of other feeding related signals and receptors expressed by VAN. In addition to its satiation effects, CCK acts all along the GI tract to optimize digestion and nutrient absorption. Diet-induced obesity (DIO) is characterized by reduced sensitivity to CCK and every part of the CCK system is negatively affected by chronic intake of energy-dense foods. EEC have recently been shown to adapt to diet, CCK1R is affected by dietary fats consumption, and the VAN phenotypic flexibility is lost in DIO. Altered endocannabinoid tone, changes in gut microbiota composition, and chronic inflammation are currently being explored as potential mechanisms for diet driven loss in CCK signaling. This review discusses our current understanding of how CCK controls food intake in conditions of leanness and how control is lost in chronic energy excess and obesity, potentially perpetuating excessive intake.
Collapse
Affiliation(s)
- Carolina R Cawthon
- Department of Foods and Nutrition, University of Georgia, Athens, GA, USA
| | | |
Collapse
|
7
|
Ong ZY, McNally GP. CART in energy balance and drug addiction: Current insights and mechanisms. Brain Res 2020; 1740:146852. [DOI: 10.1016/j.brainres.2020.146852] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022]
|
8
|
Wang YB, de Lartigue G, Page AJ. Dissecting the Role of Subtypes of Gastrointestinal Vagal Afferents. Front Physiol 2020; 11:643. [PMID: 32595525 PMCID: PMC7300233 DOI: 10.3389/fphys.2020.00643] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/20/2020] [Indexed: 12/22/2022] Open
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.
Collapse
Affiliation(s)
- Yoko B Wang
- Vagal Afferent Research Group, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Guillaume de Lartigue
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States.,Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL, United States
| | - Amanda J Page
- Vagal Afferent Research Group, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.,Nutrition, Diabetes and Gut Health, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| |
Collapse
|
9
|
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, de Lartigue G. Blunted Vagal Cocaine- and Amphetamine-Regulated Transcript Promotes Hyperphagia and Weight Gain. Cell Rep 2020; 30:2028-2039.e4. [PMID: 32049029 PMCID: PMC7063787 DOI: 10.1016/j.celrep.2020.01.045] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/06/2019] [Accepted: 01/15/2020] [Indexed: 12/31/2022] Open
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.
Collapse
Affiliation(s)
- Shin J Lee
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Jean-Philippe Krieger
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland; Department of Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Macarena Vergara
- Department of Pharmacodynamics, Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL, USA
| | | | - Molly McDougle
- Department of Pharmacodynamics, Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL, USA; The John B. Pierce Laboratory, New Haven, CT, USA
| | - Alan de Araujo
- Department of Pharmacodynamics, Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL, USA; The John B. Pierce Laboratory, New Haven, CT, USA; Yale University, New Haven, CT, USA
| | - Rebecca Darling
- Anatomy, Physiology and Cell Biology Department School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Benjamin Zollinger
- The John B. Pierce Laboratory, New Haven, CT, USA; Yale University, New Haven, CT, USA
| | - Seth Anderson
- The John B. Pierce Laboratory, New Haven, CT, USA; Yale University, New Haven, CT, USA
| | - Annabeth Pan
- The John B. Pierce Laboratory, New Haven, CT, USA; Yale University, New Haven, CT, USA
| | - Emilie J Simonnet
- Anatomy, Physiology and Cell Biology Department School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Angelica Pignalosa
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Myrtha Arnold
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Arashdeep Singh
- Department of Pharmacodynamics, Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL, USA
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Helen E Raybould
- Anatomy, Physiology and Cell Biology Department School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Guillaume de Lartigue
- Department of Pharmacodynamics, Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL, USA; The John B. Pierce Laboratory, New Haven, CT, USA; Yale University, New Haven, CT, USA.
| |
Collapse
|
10
|
Cristina Oliveira de Lima V, Piuvezam G, Leal Lima Maciel B, Heloneida de Araújo Morais A. Trypsin inhibitors: promising candidate satietogenic proteins as complementary treatment for obesity and metabolic disorders? J Enzyme Inhib Med Chem 2019; 34:405-419. [PMID: 30734596 PMCID: PMC6327991 DOI: 10.1080/14756366.2018.1542387] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 10/16/2018] [Accepted: 10/26/2018] [Indexed: 10/27/2022] Open
Abstract
The increase in non-communicable chronic diseases has aroused interest in the research of adjuvants to the classic forms of treatments. Obesity and metabolic syndrome are the main targets of confrontation because they relate directly to other chronic diseases. In this context, trypsin inhibitors, molecules with wide heterologous application, appear as possibilities in the treatment of overweight and obesity due to the action on satiety related mechanisms, mainly in the modulation of satiety hormones, such as cholecystokinin. In addition, trypsin inhibitors have the ability to also act on some biochemical parameters related to these diseases, thus, emerging as potential candidates and promising molecules in the treatment of the obesity and metabolic syndrome. Thus, the present article proposes to approach, through a systematic literature review, the advantages, disadvantages and viabilities for the use of trypsin inhibitors directed to the treatment of overweight and obesity.
Collapse
Affiliation(s)
| | - Grasiela Piuvezam
- Department of Collective Health, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Bruna Leal Lima Maciel
- Department of Nutrition, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Ana Heloneida de Araújo Morais
- Department of Biochemistry, Biosciences Center, Federal University of Rio Grande do Norte, Natal, Brazil
- Department of Nutrition, Center for Health Sciences, Federal University of Rio Grande do Norte, Natal, Brazil
| |
Collapse
|
11
|
Langhans W. Serendipity and spontaneity - Critical components in 40 years of academia. Physiol Behav 2019; 204:76-85. [PMID: 30753847 DOI: 10.1016/j.physbeh.2019.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 02/08/2019] [Indexed: 10/27/2022]
Abstract
I was flattered and felt tremendously honored to receive the 2018 Distinguished Career Award (DCA) from SSIB, the society that I always considered my scientific home, my family. Preparing the award lecture, I reflected about defining features of my career. This paper summarizes this very personal retrospective. As you will read, serendipity and more or less spontaneous decisions; i.e., some luck to be in the right place at the right time, and spontaneity to grab an opportunity when it presented itself, played a major role, and not necessarily a thorough analysis of my life situation at various junctions of my career path. Luck also often had the name of a fantastic tutor or mentor, or came in the form of enlightening discussions with a friend. Science is teamwork, which emphasizes how important collaborators, post-docs, students and technicians are. Although deep thinking was not necessarily crucial for my career path, a thorough examination is of course necessary when analyzing data, which were often most important when they did not confirm my hypothesis. Science is also hard work considering how much time one spends, but it never seemed like work to me because I had always this desire to find out how things in the organism work, and I always felt privileged to be able to pursue my "hobby" and even get a decent pay for it. In short, being a scientist is probably one of the most rewarding professional activities that life can offer.
Collapse
Affiliation(s)
- Wolfgang Langhans
- Physiology and Behavior Laboratory, Department of Health Sciences and Technology, ETH Zurich, Schorenstr. 16, 8603 Schwerzenbach, Switzerland.
| |
Collapse
|
12
|
Lai CC, Yuan ZF, Chu LY, Chuang KT, Lin HH. Roles of cocaine- and amphetamine-regulated transcript peptide in the rostral ventrolateral medulla in cardiovascular regulation in rats. Brain Res 2019; 1710:117-124. [PMID: 30610873 DOI: 10.1016/j.brainres.2019.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/22/2018] [Accepted: 01/01/2019] [Indexed: 02/07/2023]
Abstract
Cocaine- and amphetamine-regulated transcript peptide (CARTp) is present in neurons and varicose fibers in the rostral ventrolateral medulla (RVLM) that is crucial in the control of cardiovascular function. Prior research indicated that intracisternal administration of CARTp evokes hypertension and accumulation of Fos in the RVLM. Despite the interaction among CARTp, cardiovascular effect, and the RVLM, no studies have directly examined whether CARTp participates in cardiovascular regulation in the RVLM. The current study directly examined the modulation of blood pressure and baroreflex sensitivity by CARTp in the RVLM in the different strain of rats. Immunohistochemical study showed that CARTp immunoreactive (CART-IR) cell bodies and varicose CART-IR fibers were observed throughout the RVLM in the SD, WKY, and SHRs. Varicose CART-IR nerve fibers were particularly abundant in the WKY and SHRs. Bilateral microinjection of CARTp (30 pmol) into the RVLM caused a significant increase in mean arterial pressure (MAP) in WKY and SHRs. Bilateral microinjection of CARTp antibody (1:5000) into the RVLM displayed a fall in the basal level of the MAP in SHRs but had no effects in WKY rats. In SD rats, bilateral microinjection of CARTp (6, 30 or 60 pmol) into the RVLM did not change the MAP but attenuated phenylephrine-induced bradycardia in a dose-dependent manner. We propose that CARTp acting in the RVLM may involvement in the cardiovascular regulation either by increases in the blood pressure or by decreases in the baroreflex sensitivity in rats. Moreover, endogenous CARTp in the RVLM is associated with the maintenance of basal blood pressure of SHRs.
Collapse
Affiliation(s)
- Chih-Chia Lai
- Department of Pharmacology, School of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Zung Fan Yuan
- Department of Physiology, School of Medicine, Tzu Chi University, Hualien 970, Taiwan; Master Program in Medical Physiology, School of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Ling-Ying Chu
- Master Program in Medical Physiology, School of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Kai-Tung Chuang
- Master Program in Medical Physiology, School of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Hsun-Hsun Lin
- Department of Physiology, School of Medicine, Tzu Chi University, Hualien 970, Taiwan; Master Program in Medical Physiology, School of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| |
Collapse
|
13
|
Ahmadian-Moghadam H, Sadat-Shirazi MS, Zarrindast MR. Cocaine- and amphetamine-regulated transcript (CART): A multifaceted neuropeptide. Peptides 2018; 110:56-77. [PMID: 30391426 DOI: 10.1016/j.peptides.2018.10.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 10/15/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022]
Abstract
Over the last 35 years, the continuous discovery of novel neuropeptides has been the key to the better understanding of how the central nervous system has integrated with neuronal signals and behavioral responses. Cocaine and amphetamine-regulated transcript (CART) was discovered in 1995 in the rat striatum but later was found to be highly expressed in the hypothalamus. The widespread distribution of CART peptide in the brain complicated the understanding of the role played by this neurotransmitter. The main objective of the current compact review is to piece together the fragments of available information about origin, expression, distribution, projection, and function of CART peptides. Accumulative evidence suggests CART as a neurotransmitter and neuroprotective agent that is mainly involved in regulation of feeding, addiction, stress, anxiety, innate fear, neurological disease, neuropathic pain, depression, osteoporosis, insulin secretion, learning, memory, reproduction, vision, sleep, thirst and body temperature. In spite of the vast number of studies about the CART, the overall pictures about the CART functions are sketchy. First, there is a lack of information about cloned receptor, specific agonist and antagonist. Second, CART peptides are detected in discrete sets of neurons that can modulate countless activities and third; CART peptides exist in several fragments due to post-translational processing. For these reasons the overall picture about the CART peptides are sketchy and confounding.
Collapse
Affiliation(s)
- Hamid Ahmadian-Moghadam
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mohammad-Reza Zarrindast
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Endocrinology and Metabolism Research Institute, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
14
|
Abstract
The regulation of energy and glucose balance contributes to whole-body metabolic homeostasis, and such metabolic regulation is disrupted in obesity and diabetes. Metabolic homeostasis is orchestrated partly in response to nutrient and vagal-dependent gut-initiated functions. Specifically, the sensory and motor fibres of the vagus nerve transmit intestinal signals to the central nervous system and exert biological and physiological responses. In the past decade, the understanding of the regulation of vagal afferent signals and of the associated metabolic effect on whole-body energy and glucose balance has progressed. This Review highlights the contributions made to the understanding of the vagal afferent system and examines the integrative role of the vagal afferent in gastrointestinal regulation of appetite and glucose homeostasis. Investigating the integrative and metabolic role of vagal afferent signalling represents a potential strategy to discover novel therapeutic targets to restore energy and glucose balance in diabetes and obesity.
Collapse
|
15
|
Ueta Y. [Nutrient Sensing and Anorexia via Neuropeptides]. YAKUGAKU ZASSHI 2018; 138:1017-1024. [PMID: 30068841 DOI: 10.1248/yakushi.17-00048-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Various neuropeptides play an essential role in the nutrient sensing mechanism and related homeostasis. Nesfatin-1 is a newly identified neuropeptide having anorectic activity, and nesfatin-1-containing neurons are widely distributed in the brain, including the hypothalamus and brain stem. Our previous study showed that dehydration-induced anorectic effects are mediated via the central nesfatin-1 pathway in rats. Our recent studies have also shown that peripheral anorectic peptides (cholecystokinin-8, glucagon-like peptide-1, and leptin) and an antineoplastic agent (cisplatin) caused inhibition of feeding via the central nesfatin-1 pathway in rats. Nesfatin-1-containing neurons in the central nervous system, in particular the hypothalamus and the brain stem, may mediate peripheral nutrient signals and regulate feeding behavior.
Collapse
Affiliation(s)
- Yoichi Ueta
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health
| |
Collapse
|
16
|
Levels of Cocaine- and Amphetamine-Regulated Transcript in Vagal Afferents in the Mouse Are Unaltered in Response to Metabolic Challenges. eNeuro 2016; 3:eN-FTR-0174-16. [PMID: 27822503 PMCID: PMC5088776 DOI: 10.1523/eneuro.0174-16.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/15/2016] [Accepted: 09/16/2016] [Indexed: 12/21/2022] Open
Abstract
Cocaine- and amphetamine-regulated transcript (CART) is one of the most abundant neuropeptides in vagal afferents, including those involved in regulating feeding. Recent observations indicate that metabolic challenges dramatically alter the neuropeptidergic profile of CART-producing vagal afferents. Here, using confocal microscopy, we reassessed the distribution and regulation of CART(55–102) immunoreactivity in vagal afferents of the male mouse in response to metabolic challenges, including fasting and high-fat-diet feeding. Importantly, the perikarya and axons of vagal C-fibers were labeled using mice expressing channelrodhopsin-2 (ChR2-YFP) in Nav1.8-Cre–expressing neurons. In these mice, approximately 82% of the nodose ganglion neurons were labeled with ChR2-YFP. Furthermore, ChR2-YFP–labeled axons could easily be identified in the dorsovagal complex. CART(55–102) immunoreactivity was observed in 55% of the ChR2-YFP–labeled neurons in the nodose ganglion and 22% of the ChR2-YFP–labeled varicosities within the area postrema of fed, fasted, and obese mice. The distribution of positive profiles was also identical across the full range of CART staining in fed, fasted, and obese mice. In contrast to previous studies, fasting did not induce melanin-concentrating hormone (MCH) immunoreactivity in vagal afferents. Moreover, prepro-MCH mRNA was undetectable in the nodose ganglion of fasted mice. In summary, this study showed that the perikarya and central terminals of vagal afferents are invariably enriched in CART and devoid of MCH.
Collapse
|
17
|
de Lartigue G. Role of the vagus nerve in the development and treatment of diet-induced obesity. J Physiol 2016; 594:5791-5815. [PMID: 26959077 DOI: 10.1113/jp271538] [Citation(s) in RCA: 164] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/26/2016] [Indexed: 12/21/2022] Open
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.
Collapse
Affiliation(s)
- Guillaume de Lartigue
- The John B. Pierce Laboratory, New Haven, CT, USA. .,Dept Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
18
|
González-Arancibia C, Escobar-Luna J, Barrera-Bugueño C, Díaz-Zepeda C, González-Toro MP, Olavarría-Ramírez L, Zanelli-Massai F, Gotteland M, Bravo JA, Julio-Pieper M. What goes around comes around: novel pharmacological targets in the gut-brain axis. Therap Adv Gastroenterol 2016; 9:339-53. [PMID: 27134664 PMCID: PMC4830101 DOI: 10.1177/1756283x16630718] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The gut and the brain communicate bidirectionally through anatomic and humoral pathways, establishing what is known as the gut-brain axis. Therefore, interventions affecting one system will impact on the other, giving the opportunity to investigate and develop future therapeutic strategies that target both systems. Alterations in the gut-brain axis may arise as a consequence of changes in microbiota composition (dysbiosis), modifications in intestinal barrier function, impairment of enteric nervous system, unbalanced local immune response and exaggerated responses to stress, to mention a few. In this review we analyze and discuss several novel pharmacological targets within the gut-brain axis, with potential applications to improve intestinal and mental health.
Collapse
Affiliation(s)
- Camila González-Arancibia
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Jorge Escobar-Luna
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Camila Barrera-Bugueño
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Camilo Díaz-Zepeda
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - María P. González-Toro
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Loreto Olavarría-Ramírez
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Francesca Zanelli-Massai
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Martin Gotteland
- Departamento de Nutrición, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Javier A. Bravo
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | | |
Collapse
|
19
|
Iwasaki Y, Maejima Y, Suyama S, Yoshida M, Arai T, Katsurada K, Kumari P, Nakabayashi H, Kakei M, Yada T. Peripheral oxytocin activates vagal afferent neurons to suppress feeding in normal and leptin-resistant mice: a route for ameliorating hyperphagia and obesity. Am J Physiol Regul Integr Comp Physiol 2015; 308:R360-9. [DOI: 10.1152/ajpregu.00344.2014] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Oxytocin (Oxt), a neuropeptide produced in the hypothalamus, is implicated in regulation of feeding. Recent studies have shown that peripheral administration of Oxt suppresses feeding and, when infused subchronically, ameliorates hyperphagic obesity. However, the route through which peripheral Oxt informs the brain is obscure. This study aimed to explore whether vagal afferents mediate the sensing and anorexigenic effect of peripherally injected Oxt in mice. Intraperitoneal Oxt injection suppressed food intake and increased c-Fos expression in nucleus tractus solitarius to which vagal afferents project. The Oxt-induced feeding suppression and c-Fos expression in nucleus tractus solitarius were blunted in mice whose vagal afferent nerves were blocked by subdiaphragmatic vagotomy or capsaicin treatment. Oxt induced membrane depolarization and increases in cytosolic Ca2+ concentration ([Ca2+]i) in single vagal afferent neurons. The Oxt-induced [Ca2+]i increases were markedly suppressed by Oxt receptor antagonist. These Oxt-responsive neurons also responded to cholecystokinin-8 and contained cocaine- and amphetamine-regulated transcript. In obese diabetic db/db mice, leptin failed to increase, but Oxt increased [Ca2+]i in vagal afferent neurons, and single or subchronic infusion of Oxt decreased food intake and body weight gain. These results demonstrate that peripheral Oxt injection suppresses food intake by activating vagal afferent neurons and thereby ameliorates obesity in leptin-resistant db/db mice. The peripheral Oxt-regulated vagal afferent neuron provides a novel target for treating hyperphagia and obesity.
Collapse
Affiliation(s)
- Yusaku Iwasaki
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Yuko Maejima
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Shigetomo Suyama
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Masashi Yoshida
- First Department of Medicine, Saitama Medical Center, Jichi Medical University School of Medicine, Saitama, Japan
| | - Takeshi Arai
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Kenichi Katsurada
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Parmila Kumari
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Hajime Nakabayashi
- Health Science Service Center, Kanazawa University, Ishikawa, Japan; and
| | - Masafumi Kakei
- First Department of Medicine, Saitama Medical Center, Jichi Medical University School of Medicine, Saitama, Japan
| | - Toshihiko Yada
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Tochigi, Japan
- Division of Adaptation Development, Department of Developmental Physiology, National Institute for Physiological Sciences, Aichi, Japan
| |
Collapse
|
20
|
Lau J, Herzog H. CART in the regulation of appetite and energy homeostasis. Front Neurosci 2014; 8:313. [PMID: 25352770 PMCID: PMC4195273 DOI: 10.3389/fnins.2014.00313] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/17/2014] [Indexed: 12/20/2022] Open
Abstract
The cocaine- and amphetamine-regulated transcript (CART) has been the subject of significant interest for over a decade. Work to decipher the detailed mechanism of CART function has been hampered by the lack of specific pharmacological tools like antagonists and the absence of a specific CART receptor(s). However, extensive research has been devoted to elucidate the role of the CART peptide and it is now evident that CART is a key neurotransmitter and hormone involved in the regulation of diverse biological processes, including food intake, maintenance of body weight, reward and addiction, stress response, psychostimulant effects and endocrine functions (Rogge et al., 2008; Subhedar et al., 2014). In this review, we focus on knowledge gained on CART's role in controlling appetite and energy homeostasis, and also address certain species differences between rodents and humans.
Collapse
Affiliation(s)
- Jackie Lau
- Neuroscience Division, Garvan Institute of Medical Research Sydney, NSW, Australia
| | - Herbert Herzog
- Neuroscience Division, Garvan Institute of Medical Research Sydney, NSW, Australia
| |
Collapse
|
21
|
de Lartigue G, Ronveaux CC, Raybould HE. Deletion of leptin signaling in vagal afferent neurons results in hyperphagia and obesity. Mol Metab 2014; 3:595-607. [PMID: 25161883 PMCID: PMC4142400 DOI: 10.1016/j.molmet.2014.06.003] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 06/18/2014] [Accepted: 06/21/2014] [Indexed: 12/15/2022] Open
Abstract
The vagal afferent pathway senses hormones released from the gut in response to nutritional cues and relays these signals to the brain. We tested the hypothesis that leptin resistance in vagal afferent neurons (VAN) is responsible for the onset of hyperphagia by developing a novel conditional knockout mouse to delete leptin receptor selectively in sensory neurons (Nav1.8/LepR (fl/fl) mice). Chow fed Nav1.8/LepR (fl/fl) mice weighed significantly more and had increased adiposity compared with wildtype mice. Cumulative food intake, meal size, and meal duration in the dark phase were increased in Nav1.8/LepR (fl/fl) mice; energy expenditure was unaltered. Reduced satiation in Nav1.8/LepR (fl/fl) mice is in part due to reduced sensitivity of VAN to CCK and the subsequent loss of VAN plasticity. Crucially Nav1.8/LepR (l/fl) mice did not gain further weight in response to a high fat diet. We conclude that disruption of leptin signaling in VAN is sufficient and necessary to promote hyperphagia and obesity.
Collapse
Affiliation(s)
- Guillaume de Lartigue
- Department of Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, 1 Shields Ave, Davis, CA 95616, USA
| | - Charlotte C Ronveaux
- Department of Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, 1 Shields Ave, Davis, CA 95616, USA
| | - Helen E Raybould
- Department of Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, 1 Shields Ave, Davis, CA 95616, USA
| |
Collapse
|
22
|
Subhedar NK, Nakhate KT, Upadhya MA, Kokare DM. CART in the brain of vertebrates: circuits, functions and evolution. Peptides 2014; 54:108-30. [PMID: 24468550 DOI: 10.1016/j.peptides.2014.01.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 01/10/2014] [Accepted: 01/10/2014] [Indexed: 12/12/2022]
Abstract
Cocaine- and amphetamine-regulated transcript peptide (CART) with its wide distribution in the brain of mammals has been the focus of considerable research in recent years. Last two decades have witnessed a steady rise in the information on the genes that encode this neuropeptide and regulation of its transcription and translation. CART is highly enriched in the hypothalamic nuclei and its relevance to energy homeostasis and neuroendocrine control has been understood in great details. However, the occurrence of this peptide in a range of diverse circuitries for sensory, motor, vegetative, limbic and higher cortical areas has been confounding. Evidence that CART peptide may have role in addiction, pain, reward, learning and memory, cognition, sleep, reproduction and development, modulation of behavior and regulation of autonomic nervous system are accumulating, but an integration has been missing. A steady stream of papers has been pointing at the therapeutic potentials of CART. The current review is an attempt at piecing together the fragments of available information, and seeks meaning out of the CART elements in their anatomical niche. We try to put together the CART containing neuronal circuitries that have been conclusively demonstrated as well as those which have been proposed, but need confirmation. With a view to finding out the evolutionary antecedents, we visit the CART systems in sub-mammalian vertebrates and seek the answer why the system is shaped the way it is. We enquire into the conservation of the CART system and appreciate its functional diversity across the phyla.
Collapse
Affiliation(s)
- Nishikant K Subhedar
- Indian Institute of Science Education and Research (IISER), Sai Trinity Building, Sutarwadi, Pashan, Pune 411 021, Maharashtra, India.
| | - Kartik T Nakhate
- Rungta College of Pharmaceutical Sciences and Research, Rungta Educational Campus, Kohka-Kurud Road, Bhilai 490 024, Chhattisgarh, India
| | - Manoj A Upadhya
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440 033, Maharashtra, India
| | - Dadasaheb M Kokare
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440 033, Maharashtra, India
| |
Collapse
|
23
|
Plasticity of gastro-intestinal vagal afferent endings. Physiol Behav 2014; 136:170-8. [PMID: 24657740 DOI: 10.1016/j.physbeh.2014.03.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Revised: 02/06/2014] [Accepted: 03/10/2014] [Indexed: 12/15/2022]
Abstract
Vagal afferents are a vital link between the peripheral tissue and central nervous system (CNS). There is an abundance of vagal afferents present within the proximal gastrointestinal tract which are responsible for monitoring and controlling gastrointestinal function. Whilst essential for maintaining homeostasis there is a vast amount of literature emerging which describes remarkable plasticity of vagal afferents in response to endogenous as well as exogenous stimuli. This plasticity for the most part is vital in maintaining healthy processes; however, there are increased reports of vagal plasticity being disrupted in pathological states, such as obesity. Many of the disruptions, observed in obesity, have the potential to reduce vagal afferent satiety signalling which could ultimately perpetuate the obese state. Understanding how plasticity occurs within vagal afferents will open a whole new understanding of gut function as well as identify new treatment options for obesity.
Collapse
|
24
|
de Lartigue G. Putative roles of neuropeptides in vagal afferent signaling. Physiol Behav 2014; 136:155-69. [PMID: 24650553 DOI: 10.1016/j.physbeh.2014.03.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 02/23/2014] [Accepted: 03/10/2014] [Indexed: 02/06/2023]
Abstract
The vagus nerve is a major pathway by which information is communicated between the brain and peripheral organs. Sensory neurons of the vagus are located in the nodose ganglia. These vagal afferent neurons innervate the heart, the lung and the gastrointestinal tract, and convey information about peripheral signals to the brain important in the control of cardiovascular tone, respiratory tone, and satiation, respectively. Glutamate is thought to be the primary neurotransmitter involved in conveying all of this information to the brain. It remains unclear how a single neurotransmitter can regulate such an extensive list of physiological functions from a wide range of visceral sites. Many neurotransmitters have been identified in vagal afferent neurons and have been suggested to modulate the physiological functions of glutamate. Specifically, the anorectic peptide transmitters, cocaine and amphetamine regulated transcript (CART) and the orexigenic peptide transmitters, melanin concentrating hormone (MCH) are differentially regulated in vagal afferent neurons and have opposing effects on food intake. Using these two peptides as a model, this review will discuss the potential role of peptide transmitters in providing a more precise and refined modulatory control of the broad physiological functions of glutamate, especially in relation to the control of feeding.
Collapse
Affiliation(s)
- Guillaume de Lartigue
- Dept Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, Davis, CA 95616, USA.
| |
Collapse
|
25
|
Dockray GJ. Enteroendocrine cell signalling via the vagus nerve. Curr Opin Pharmacol 2013; 13:954-8. [DOI: 10.1016/j.coph.2013.09.007] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 08/28/2013] [Accepted: 09/04/2013] [Indexed: 02/06/2023]
|
26
|
Pituitary adenylate cyclase-activating polypeptide 6-38 blocks cocaine- and amphetamine-regulated transcript Peptide-induced hypophagia in rats. PLoS One 2013; 8:e72347. [PMID: 23967296 PMCID: PMC3744533 DOI: 10.1371/journal.pone.0072347] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 07/08/2013] [Indexed: 02/01/2023] Open
Abstract
Cocaine- and amphetamine-regulated transcript peptides (CARTp) suppress nutritional intake after administration into the fourth intracerebral ventricle. Recent in vitro studies have shown that PACAP 6-38, a pituitary adenylate cyclase-activating polypeptide (PACAP) fragment, could act as a competitive antagonist against CARTp 55-102 on a common CARTp-sensitive receptor structure. Here, we show for the first time in vivo that the reduction in solid food intake induced by exogenous CARTp 55-102 (0.3 nmol: 1.5 µg) administered fourth i.c.v. is blocked by pretreatment with PACAP 6-38 (3 nmol). The PACAP 6-38 fragment had no effect by itself either when given into the fourth ventricle or subcutaneously. Although effective to block the CARTp-effect on feeding and short-term body weight, PACAP 6-38 failed to attenuate CARTp-associated gross motor behavioral changes suggesting at least two CARTp-sensitive receptor subtypes. In conclusion, PACAP 6-38 acts as a functional CARTp antagonist in vivo and blocks its effects on feeding and short term weight gain.
Collapse
|
27
|
Campos CA, Shiina H, Silvas M, Page S, Ritter RC. Vagal afferent NMDA receptors modulate CCK-induced reduction of food intake through synapsin I phosphorylation in adult male rats. Endocrinology 2013; 154:2613-25. [PMID: 23715865 PMCID: PMC3713210 DOI: 10.1210/en.2013-1062] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Vagal afferent nerve fibers transmit gastrointestinal satiation signals to the brain via synapses in the nucleus of the solitary tract (NTS). Despite their pivotal role in energy homeostasis, little is known about the cellular mechanisms enabling fleeting synaptic events at vagal sensory endings to sustain behavioral changes lasting minutes to hours. Previous reports suggest that the reduction of food intake by the satiation peptide, cholecystokinin (CCK), requires activation of N-methyl-D-aspartate-type glutamate receptors (NMDAR) in the NTS, with subsequent phosphorylation of ERK1/2 (pERK1/2) in NTS vagal afferent terminals. The synaptic vesicle protein synapsin I is phosphorylated by pERK1/2 at serines 62 and 67. This pERK1/2-catalyzed phosphorylation increases synaptic strength by increasing the readily releasable pool of the neurotransmitter. Conversely, dephosphorylation of serines 62 and 67 by calcineurin reduces the size of the readily releasable transmitter pool. Hence, the balance of synapsin I phosphorylation and dephosphorylation can modulate synaptic strength. We postulated that CCK-evoked activation of vagal afferent NMDARs results in pERK1/2-catalyzed phosphorylation of synapsin I in vagal afferent terminals, leading to the suppression of food intake. We found that CCK injection increased the phosphorylation of synapsin I in the NTS and that this increase is abolished after surgical or chemical ablation of vagal afferent fibers. Furthermore, fourth ventricle injection of an NMDAR antagonist or the mitogen-activated ERK kinase inhibitor blocked CCK-induced synapsin I phosphorylation, indicating that synapsin phosphorylation in vagal afferent terminals depends on NMDAR activation and ERK1/2 phosphorylation. Finally, hindbrain inhibition of calcineurin enhanced and prolonged synapsin I phosphorylation and potentiated reduction of food intake by CCK. Our findings are consistent with a mechanism in which NMDAR-dependent phosphorylation of ERK1/2 modulates satiation signals via synapsin I phosphorylation in vagal afferent endings.
Collapse
Affiliation(s)
- Carlos A Campos
- Department of Integrative Physiology and Neuroscience, Washington State University, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA.
| | | | | | | | | |
Collapse
|
28
|
Insulin Activates Vagal Afferent Neurons Including those Innervating Pancreas via Insulin Cascade and Ca(2+) Influx: Its Dysfunction in IRS2-KO Mice with Hyperphagic Obesity. PLoS One 2013; 8:e67198. [PMID: 23840624 PMCID: PMC3693960 DOI: 10.1371/journal.pone.0067198] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 05/15/2013] [Indexed: 11/19/2022] Open
Abstract
Some of insulin’s functions, including glucose/lipid metabolism, satiety and neuroprotection, involve the alteration of brain activities. Insulin could signal to the brain via penetrating through the blood-brain barrier and acting on the vagal afferents, while the latter remains unproved. This study aimed to clarify whether insulin directly regulates the nodose ganglion neurons (NGNs) of vagal afferents in mice. NGs expressed insulin receptor (IR) and insulin receptor substrate-2 (IRS2) mRNA, and some of NGNs were immunoreactive to IR. In patch-clamp and fura-2 microfluorometric studies, insulin (10−12∼10−6 M) depolarized and increased cytosolic Ca2+ concentration ([Ca2+]i) in single NGNs. The insulin-induced [Ca2+]i increases were attenuated by L- and N-type Ca2+ channel blockers, by phosphatidylinositol 3 kinase (PI3K) inhibitor, and in NGNs from IRS2 knockout mice. Half of the insulin-responsive NGNs contained cocaine- and amphetamine-regulated transcript. Neuronal fibers expressing IRs were distributed in/around pancreatic islets. The NGNs innervating the pancreas, identified by injecting retrograde tracer into the pancreas, responded to insulin with much greater incidence than unlabeled NGNs. Insulin concentrations measured in pancreatic vein was 64-fold higher than that in circulation. Elevation of insulin to 10−7 M recruited a remarkably greater population of NGNs to [Ca2+]i increases. Systemic injection of glibenclamide rapidly released insulin and phosphorylated AKT in NGs. Furthermore, in IRS2 knockout mice, insulin action to suppress [Ca2+]i in orexigenic ghrelin-responsive neurons in hypothalamic arcuate nucleus was intact while insulin action on NGN was markedly attenuated, suggesting a possible link between impaired insulin sensing by NGNs and hyperphagic obese phenotype in IRS2 knockout mice These data demonstrate that insulin directly activates NGNs via IR-IRS2-PI3K-AKT-cascade and depolarization-gated Ca2+ influx. Pancreas-innervating NGNs may effectively sense dynamic changes of insulin released in response to nutritional states. These interactions could serve to convey the changes in pancreatic and systemic insulin to the brain.
Collapse
|
29
|
Gautron L, Lee CE, Lee S, Elmquist JK. Melanocortin-4 receptor expression in different classes of spinal and vagal primary afferent neurons in the mouse. J Comp Neurol 2013; 520:3933-48. [PMID: 22592759 DOI: 10.1002/cne.23137] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Melanocortin-4 receptor (MC4R) ligands are known to modulate nociception, but the site of action of MC4R signaling on nociception remains to be elucidated. The current study investigated MC4R expression in dorsal root ganglia (DRG) of the MC4R-GFP reporter mouse. Because MC4R is known to be expressed in vagal afferent neurons in the nodose ganglion (NG), we also systematically compared MC4R-expressing vagal and spinal afferent neurons. Abundant green fluorescent protein (GFP) immunoreactivity was found in about 45% of DRG neuronal profiles (at the mid-thoracic level), the majority being small-sized profiles. Immunohistochemistry combined with in situ hybridization confirmed that GFP was genuinely produced in MC4R-expressing neurons in the DRG. While a large number of GFP profiles in the DRG coexpressed Nav1.8 mRNA (84%) and bound isolectin B4 (72%), relatively few GFP profiles were positive for NF200 (16%) or CGRP (13%), suggesting preferential MC4R expression in C-fiber nonpeptidergic neurons. By contrast, GFP in the NG frequently colocalized with Nav1.8 mRNA (64%) and NF200 (29%), but only to a moderate extent with isolectin B4 (16%). Lastly, very few GFP profiles in the NG expressed CGRP (5%) or CART (4%). Together, our findings demonstrate variegated MC4R expression in different classes of vagal and spinal primary afferent neurons, and underscore the role of the melanocortin system in modulating nociceptive and nonnociceptive peripheral sensory modalities.
Collapse
Affiliation(s)
- Laurent Gautron
- Department of Internal Medicine, Division of Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9077, USA.
| | | | | | | |
Collapse
|
30
|
Iwasaki Y, Yada T. Vagal afferents sense meal-associated gastrointestinal and pancreatic hormones: mechanism and physiological role. Neuropeptides 2012; 46:291-7. [PMID: 23020951 DOI: 10.1016/j.npep.2012.08.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 08/17/2012] [Accepted: 08/17/2012] [Indexed: 01/08/2023]
Abstract
Some gastrointestinal and pancreatic hormones are potently secreted by meal intake and reduce food intake, therefore these hormones play a role in the meal-evoked satiety peptides. Previous reports have demonstrated that peripheral administration of these gastrointestinal or pancreatic hormones decrease feeding and the anorectic effects are abolished by lesions of vagal afferent nerves using surgical or chemical protocols, indicative of the involvement of the vagal afferents. Vagal afferent nerves link between several peripheral organs and the nucleus tractus solitarius of the brainstem. The present review focuses on cholecystokinin, peptide YY(3-36), pancreatic polypeptide, and nesfatin-1 released from endocrine cells of the gut and pancreas. These hormonal peptides directly act on and increase cytosolic Ca(2+) in vagal afferent nodose ganglion neurons and finally suppress food intake via vagal afferents. Therefore, peripheral terminals of vagal afferents could sense gastrointestinal and pancreatic hormones and regulate food intake. Here, we review how the vagal afferent neurons sense a variety of gastrointestinal and pancreatic hormones and discuss its physiological significance in regulation of feeding.
Collapse
Affiliation(s)
- Yusaku Iwasaki
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, 3311-1, Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | | |
Collapse
|
31
|
Heldsinger A, Lu Y, Zhou SY, Wu X, Grabauskas G, Song I, Owyang C. Cocaine- and amphetamine-regulated transcript is the neurotransmitter regulating the action of cholecystokinin and leptin on short-term satiety in rats. Am J Physiol Gastrointest Liver Physiol 2012; 303:G1042-51. [PMID: 22936273 PMCID: PMC3517666 DOI: 10.1152/ajpgi.00231.2012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Vagal CCK-A receptors (CCKARs) and leptin receptors (LRbs) interact synergistically to mediate short-term satiety. Cocaine- and amphetamine-regulated transcript (CART) peptide is expressed by vagal afferent neurons. We sought to demonstrate that this neurotransmitter regulates CCK and leptin actions on short-term satiety. We also examined the signal transduction pathways responsible for mediating the CART release from the nodose ganglia (NG). ELISA studies coupled with gene silencing of NG neurons by RNA interference elucidated intracellular signaling pathways responsible for CCK/leptin-stimulated CART release. Feeding studies followed by gene silencing of CART in NG established the role of CART in mediating short-term satiety. Immunohistochemistry was performed on rat NG neurons to confirm colocalization of CCKARs and LRbs; 63% of these neurons contained CART. Coadministration of CCK-8 and leptin caused a 2.2-fold increase in CART release that was inhibited by CCK-OPE, a low-affinity CCKAR antagonist. Transfection of cultured NG neurons with steroid receptor coactivator (SRC) or phosphatidylinositol 3-kinase (PI3K) small-interfering RNA (siRNA) or STAT3 lentiviral short hairpin RNA inhibited CCK/leptin-stimulated CART release. Silencing the expression of the EGR-1 gene inhibited the CCK/leptin-stimulated CART release but had no effect on CCK/leptin-stimulated neuronal firing. Electroporation of NG with CART siRNA inhibited CCK/leptin stimulated c-Fos expression in rat hypothalamus. Feeding studies following electroporation of the NG with CART or STAT3 siRNA abolished the effects of CCK/leptin on short-term satiety. We conclude that the synergistic interaction of low-affinity vagal CCKARs and LRbs mediates CART release from the NG, and CART is the principal neurotransmitter mediating short-term satiety. CART release from the NG involves interaction between CCK/SRC/PI3K cascades and leptin/JAK2/PI3K/STAT3 signaling pathways.
Collapse
Affiliation(s)
- Andrea Heldsinger
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Yuanxu Lu
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Shi-Yi Zhou
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Xiaoyin Wu
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Gintautas Grabauskas
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Il Song
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Chung Owyang
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
32
|
Page AJ, Symonds E, Peiris M, Blackshaw LA, Young RL. Peripheral neural targets in obesity. Br J Pharmacol 2012; 166:1537-58. [PMID: 22432806 PMCID: PMC3419899 DOI: 10.1111/j.1476-5381.2012.01951.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Revised: 02/20/2012] [Accepted: 02/22/2012] [Indexed: 12/15/2022] Open
Abstract
Interest in pharmacological treatments for obesity that act in the brain to reduce appetite has increased exponentially over recent years, but failures of clinical trials and withdrawals due to adverse effects have so far precluded any success. Treatments that do not act within the brain are, in contrast, a neglected area of research and development. This is despite the fact that a vast wealth of molecular mechanisms exists within the gut epithelium and vagal afferent system that could be manipulated to increase satiety. Here we discuss mechano- and chemosensory pathways from the gut involved in appetite suppression, and distinguish between gastric and intestinal vagal afferent pathways in terms of their basic physiology and activation by enteroendocrine factors. Gastric bypass surgery makes use of this system by exposing areas of the intestine to greater nutrient loads resulting in greater satiety hormone release and reduced food intake. A non-surgical approach to this system is preferable for many reasons. This review details where the opportunities may lie for such approaches by describing nutrient-sensing mechanisms throughout the gastrointestinal tract.
Collapse
Affiliation(s)
- Amanda J Page
- Nerve-Gut Research Laboratory, Discipline of Medicine, South Australia, Australia
| | | | | | | | | |
Collapse
|
33
|
de Lartigue G, Barbier de la Serre C, Espero E, Lee J, Raybould HE. Leptin resistance in vagal afferent neurons inhibits cholecystokinin signaling and satiation in diet induced obese rats. PLoS One 2012; 7:e32967. [PMID: 22412960 PMCID: PMC3296757 DOI: 10.1371/journal.pone.0032967] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 02/06/2012] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND AND AIMS The gastrointestinal hormone cholecystokinin (CCK) plays an important role in regulating meal size and duration by activating CCK1 receptors on vagal afferent neurons (VAN). Leptin enhances CCK signaling in VAN via an early growth response 1 (EGR1) dependent pathway thereby increasing their sensitivity to CCK. In response to a chronic ingestion of a high fat diet, VAN develop leptin resistance and the satiating effects of CCK are reduced. We tested the hypothesis that leptin resistance in VAN is responsible for reducing CCK signaling and satiation. RESULTS Lean Zucker rats sensitive to leptin signaling, significantly reduced their food intake following administration of CCK8S (0.22 nmol/kg, i.p.), while obese Zucker rats, insensitive to leptin, did not. CCK signaling in VAN of obese Zucker rats was reduced, preventing CCK-induced up-regulation of Y2 receptor and down-regulation of melanin concentrating hormone 1 receptor (MCH1R) and cannabinoid receptor (CB1). In VAN from diet-induced obese (DIO) Sprague Dawley rats, previously shown to become leptin resistant, we demonstrated that the reduction in EGR1 expression resulted in decreased sensitivity of VAN to CCK and reduced CCK-induced inhibition of food intake. The lowered sensitivity of VAN to CCK in DIO rats resulted in a decrease in Y2 expression and increased CB1 and MCH1R expression. These effects coincided with the onset of hyperphagia in DIO rats. CONCLUSIONS Leptin signaling in VAN is required for appropriate CCK signaling and satiation. In response to high fat feeding, the onset of leptin resistance reduces the sensitivity of VAN to CCK thus reducing the satiating effects of CCK.
Collapse
Affiliation(s)
- Guillaume de Lartigue
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Claire Barbier de la Serre
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Elvis Espero
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Jennifer Lee
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Helen E. Raybould
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW Cholecystokinin (CCK) controls nutrient delivery to the small intestine by inhibiting food intake and gastric emptying. This review deals with recent work shedding new light on how and when. RECENT FINDINGS Intestinal I-cells release CCK in response to dietary lipid and protein through mechanisms involving the G-protein-coupled receptors GPR40 and calcium-sensing receptor. Vagal afferent neurons are a primary target of CCK and are now recognized as an important site of integration of peripheral signals regulating ingestion. In addition to regulating vagal afferent nerve discharge, CCK also controls the expression of receptors and peptide neurotransmitters by these neurons; these actions are potentiated by leptin and inhibited by ghrelin. The responses of vagal afferent neurons to CCK are attenuated in obesity. Studies of human central nervous system responses using functional magnetic resonance imaging indicate activation of brainstem, hypothalamus and motor cortex by ingested fatty acid that is inhibited by a CCK-1 receptor antagonist. CCK may also play a role in adaptive responses in pancreatic islets by maintaining β-cell mass and acting as an incretin in certain circumstances. SUMMARY CCK mediates inhibition of food intake in response to ingested lipid and protein; resistance to CCK occurs in obesity and may contribute to altered mechanisms regulating food intake.
Collapse
Affiliation(s)
- Graham J Dockray
- Physiological Laboratory, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| |
Collapse
|
35
|
Ivanusic JJ, Goulding KE, Kwok MMK, Jennings EA. Neurochemical classification and projection targets of CART peptide immunoreactive neurons in sensory and parasympathetic ganglia of the head. Neuropeptides 2012; 46:55-60. [PMID: 22005173 DOI: 10.1016/j.npep.2011.09.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 09/22/2011] [Accepted: 09/22/2011] [Indexed: 10/16/2022]
Abstract
The aims of the present study were to determine if there is neuronal Cocaine and amphetamine regulated transcripts (CART) peptide expression (CART+) in parasympathetic (sphenopalatine (SPG); otic (OG)) and sensory (trigeminal (TG)) ganglia of the head and to examine the neurochemical phenotype (calcitonin gene-related peptide (CGRP), neurofilament 200 (NF200), isolectin B4 (IB4) binding, vasoactive intestinal peptide (VIP), neuropeptide Y (NPY) and enkephalin (ENK) immunoreactivity) and projection targets (lacrimal gland (LG), parotid gland (PG), nasal mucosa (NM), temporomandibular joint (TMJ), middle cerebral artery (MCA) and middle meningeal artery (MMA)) of CART expressing neurons in these ganglia. We found CART+ neurons in both the SPG (5.25±0.07%) and OG (4.32±0.66). A significant proportion of these CART+ neurons contained VIP, NPY or ENK (34%, 26% and 11%, respectively). SPG neurons retrogradely labelled from the lacrimal gland (29%) were CART+, but we were unable to demonstrate CART+ labelling in any of the SPG or OG neurons labelled from other targets. This supports a role for CART peptides in lacrimation or regulation of vascular tone in the lacrimal gland, but not in salivation or nasal congestion. CART+ neurons were also present in the trigeminal ganglion (1.26±0.38%), where their size distribution was confined almost completely to neurons smaller than 800 μm2 (mean=410 μm2; 98%<800 μm2), and were almost always CGRP+, but did not bind IB4. This is consistent with a role for CART peptides in trigeminal pain. However, there were few CART+ neurons amongst any of the trigeminal neurons retrogradely labelled from the targets we investigated and thus we cannot comment on the tissue type where such pain may have originated. Our study shows that some specialization of CART peptide expression (based on neurochemical phenotype and target projection) is evident in sensory and parasympathetic ganglia of the head.
Collapse
Affiliation(s)
- Jason J Ivanusic
- Department of Anatomy & Cell Biology, University of Melbourne, Parkville 3010, Australia
| | | | | | | |
Collapse
|
36
|
Plank C, Zelphati O, Mykhaylyk O. Magnetically enhanced nucleic acid delivery. Ten years of magnetofection-progress and prospects. Adv Drug Deliv Rev 2011; 63:1300-31. [PMID: 21893135 PMCID: PMC7103316 DOI: 10.1016/j.addr.2011.08.002] [Citation(s) in RCA: 251] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 08/18/2011] [Accepted: 08/19/2011] [Indexed: 12/28/2022]
Abstract
Nucleic acids carry the building plans of living systems. As such, they can be exploited to make cells produce a desired protein, or to shut down the expression of endogenous genes or even to repair defective genes. Hence, nucleic acids are unique substances for research and therapy. To exploit their potential, they need to be delivered into cells which can be a challenging task in many respects. During the last decade, nanomagnetic methods for delivering and targeting nucleic acids have been developed, methods which are often referred to as magnetofection. In this review we summarize the progress and achievements in this field of research. We discuss magnetic formulations of vectors for nucleic acid delivery and their characterization, mechanisms of magnetofection, and the application of magnetofection in viral and nonviral nucleic acid delivery in cell culture and in animal models. We summarize results that have been obtained with using magnetofection in basic research and in preclinical animal models. Finally, we describe some of our recent work and end with some conclusions and perspectives.
Collapse
|
37
|
Wernette CM, White BD, Zizza CA. Signaling proteins that influence energy intake may affect unintentional weight loss in elderly persons. ACTA ACUST UNITED AC 2011; 111:864-73. [PMID: 21616199 DOI: 10.1016/j.jada.2011.03.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Accepted: 12/08/2010] [Indexed: 11/25/2022]
Abstract
After age 70 to 75 years, average body weight decreases both in ailing and healthy people because of a loss of appetite that results in reduced energy intake and the loss of body fat and lean muscle tissue. This so-called anorexia of aging predisposes elderly people to continued pathologic weight loss and malnutrition-major causes of morbidity and mortality. Health care professionals must understand the many factors involved in the anorexia of aging to help older adults prevent unintentional weight loss. Psychological, social, and cultural factors are important effectors; however, physiological factors are emphasized here because they are not thoroughly understood and they make it inherently difficult for most people to alter their body weight. Monoamines, steroid hormones (glucocorticoids and mineralocorticoids), endocannabinoids, and proteins all influence body weight. This review is an analysis of proteins from the brain, pancreas, adipose tissue, and gastrointestinal tract that are known to affect energy intake and energy balance, with an attempt to identify those factors that may change with aging. The articles included in this review were obtained by a PubMed database search using the keywords mouse OR rat OR human AND aged OR aging OR older OR elderly AND adult AND anorexia OR "unintentional weight loss," and each of the individual proteins discussed, as well as from the reference lists of those articles. The results reveal that some proteins may be important in the development of unintentional weight loss in elderly persons, whereas others may not have a significant role. However, many of the proteins that could conceivably have a role in unintentional weight loss have not yet been studied with that question in mind. Preventing unintentional weight loss in older adults is an important goal and further research on the role of proteins important for the maintenance of energy balance and the development of unintentional weight loss in elderly persons is warranted.
Collapse
Affiliation(s)
- Catherine M Wernette
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL, USA.
| | | | | |
Collapse
|
38
|
Abstract
The concept that the gut and the brain are closely connected, and that this interaction plays an important part not only in gastrointestinal function but also in certain feeling states and in intuitive decision making, is deeply rooted in our language. Recent neurobiological insights into this gut-brain crosstalk have revealed a complex, bidirectional communication system that not only ensures the proper maintenance of gastrointestinal homeostasis and digestion but is likely to have multiple effects on affect, motivation and higher cognitive functions, including intuitive decision making. Moreover, disturbances of this system have been implicated in a wide range of disorders, including functional and inflammatory gastrointestinal disorders, obesity and eating disorders.
Collapse
Affiliation(s)
- Emeran A Mayer
- Center for Neurobiology of Stress, Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CHS 47-122 10833 Le Conte Avenue, Los Angeles, California 90095-7378, USA.
| |
Collapse
|
39
|
de Lartigue G, Barbier de la Serre C, Espero E, Lee J, Raybould HE. Diet-induced obesity leads to the development of leptin resistance in vagal afferent neurons. Am J Physiol Endocrinol Metab 2011; 301:E187-95. [PMID: 21521717 PMCID: PMC3129833 DOI: 10.1152/ajpendo.00056.2011] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Ingestion of high-fat, high-calorie diets is associated with hyperphagia, increased body fat, and obesity. The mechanisms responsible are currently unclear; however, altered leptin signaling may be an important factor. Vagal afferent neurons (VAN) integrate signals from the gut in response to ingestion of nutrients and express leptin receptors. Therefore, we tested the hypothesis that leptin resistance occurs in VAN in response to a high-fat diet. Sprague-Dawley rats, which exhibit a bimodal distribution of body weight gain, were used after ingestion of a high-fat diet for 8 wk. Body weight, food intake, and plasma leptin levels were measured. Leptin signaling was determined by immunohistochemical localization of phosphorylated STAT3 (pSTAT3) in cultured VAN and by quantifaction of pSTAT3 protein levels by Western blot analysis in nodose ganglia and arcuate nucleus in vivo. To determine the mechanism of leptin resistance in nodose ganglia, cultured VAN were stimulated with leptin alone or with lipopolysaccharide (LPS) and SOCS-3 expression measured. SOCS-3 protein levels in VAN were measured by Western blot following leptin administration in vivo. Leptin resulted in appearance of pSTAT3 in VAN of low-fat-fed rats and rats resistant to diet-induced obesity but not diet-induced obese (DIO) rats. However, leptin signaling was normal in arcuate neurons. SOCS-3 expression was increased in VAN of DIO rats. In cultured VAN, LPS increased SOCS-3 expression and inhibited leptin-induced pSTAT3 in vivo. We conclude that VAN of diet-induced obese rats become leptin resistant; LPS and SOCS-3 may play a role in the development of leptin resistance.
Collapse
Affiliation(s)
- Guillaume de Lartigue
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | | | | | | | | |
Collapse
|
40
|
Valen R, Jordal AEO, Murashita K, Rønnestad I. Postprandial effects on appetite-related neuropeptide expression in the brain of Atlantic salmon, Salmo salar. Gen Comp Endocrinol 2011; 171:359-66. [PMID: 21377470 DOI: 10.1016/j.ygcen.2011.02.027] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Revised: 02/24/2011] [Accepted: 02/27/2011] [Indexed: 11/29/2022]
Abstract
Following feeding of a single meal to Atlantic salmon, the temporal changes in the brain mRNA expression of neuropeptide y (npy), cocaine-amphetamine regulated transcript (cart), peptide yy (pyy), two isoforms of agouti-related protein (agrp), two isoforms of cholecystokinin (cck), and four isoforms of proopiomelanocortin (pomc) were assessed by q-PCR. In the course of 24h post-feeding (hpf), several of the brain neuropeptides displayed changes in mRNA expression compared to an unfed control group, indicating that food intake and processing affect the regulation of expression of these genes in Atlantic salmon. Expression of cart, cck-l, pomc-a1 and pomc-b all increased within 3h of feeding, while most of the feed was still in the stomach, suggesting that these neuropeptides play central anorexigenic roles similar to those described in higher vertebrates, including determining meal intervals. On the other hand, the npy and agrp isoforms which have been described as playing orexigenic roles in mammals, showed an opposite response in salmon and both were elevated in the first 3h after feeding. The different isoforms of cck, agrp and pomc had different mRNA expression patterns, which indicate specific roles related to feeding regulation. The minimal effect of feeding and digestion on pyy expression in the brain indicates that PYY plays a minor role in the central control of short-term food intake in Atlantic salmon.
Collapse
Affiliation(s)
- R Valen
- Department of Biology, University of Bergen, NO-5020 Bergen, Norway.
| | | | | | | |
Collapse
|
41
|
Daly DM, Park SJ, Valinsky WC, Beyak MJ. Impaired intestinal afferent nerve satiety signalling and vagal afferent excitability in diet induced obesity in the mouse. J Physiol 2011; 589:2857-70. [PMID: 21486762 DOI: 10.1113/jphysiol.2010.204594] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Gastrointestinal vagal afferents transmit satiety signals to the brain via both chemical and mechanical mechanisms. There is indirect evidence that these signals may be attenuated in obesity. We hypothesized that responses to satiety mediators and distension of the gut would be attenuated after induction of diet induced obesity. Obesity was induced by feeding a high fat diet (60% kcal from fat). Low fat fed mice (10% kcal from fat) served as a control. High fat fed mice were obese, with increased visceral fat, but were not hyperglycaemic. Recordings from jejunal afferents demonstrated attenuated responses to the satiety mediators cholecystokinin (CCK, 100 nm) and 5-hydroxytryptamine (5-HT, 10 μm), as was the response to low intensity jejunal distension, while responses to higher distension pressures were preserved. We performed whole cell patch clamp recordings on nodose ganglion neurons, both unlabelled, and those labelled by fast blue injection into the wall of the jejunum. The cell membrane of both labelled and unlabelled nodose ganglion neurons was less excitable in HFF mice, with an elevated rheobase and decreased number of action potentials at twice rheobase. Input resistance of HFF neurons was also significantly decreased. Calcium imaging experiments revealed reduced proportion of nodose ganglion neurons responding to CCK and 5-HT in obese mice. These results demonstrate a marked reduction in afferent sensitivity to satiety related stimuli after a chronic high fat diet. A major mechanism underlying this change is reduced excitability of the neuronal cell membrane. This may explain the development of hyperphagia when a high fat diet is consumed. Improving sensitivity of gastrointestinal afferent nerves may prove useful to limit food intake in obesity.
Collapse
Affiliation(s)
- Donna M Daly
- Departments of Medicine and Physiology, Gastrointestinal Diseases Research Unit (GIDRU) Wing, Kingston General Hospital, 76 Stuart Street, Kingston, Ontario, Canada K7L2V7
| | | | | | | |
Collapse
|
42
|
de Lartigue G, de La Serre CB, Raybould HE. Vagal afferent neurons in high fat diet-induced obesity; intestinal microflora, gut inflammation and cholecystokinin. Physiol Behav 2011; 105:100-5. [PMID: 21376066 DOI: 10.1016/j.physbeh.2011.02.040] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Revised: 02/23/2011] [Accepted: 02/25/2011] [Indexed: 12/13/2022]
Abstract
The vagal afferent pathway is the major neural pathway by which information about ingested nutrients reaches the CNS and influences both GI function and feeding behavior. Vagal afferent neurons (VAN) express receptors for many of the regulatory peptides and molecules released from the intestinal wall, pancreas, and adipocytes that influence GI function, glucose homeostasis, and regulate food intake and body weight. As such, they play a critical role in both physiology and pathophysiology, such as obesity, where there is evidence that vagal afferent function is altered. This review will summarize recent findings on changes in vagal afferent function in response to ingestion of high fat diets and explore the hypothesis that changes in gut microbiota and integrity of the epithelium may not only be important in inducing these changes but may be the initial events that lead to dysregulation of food intake and body weight in response to high fat, high energy diets.
Collapse
Affiliation(s)
- Guillaume de Lartigue
- Department of Anatomy, School of Veterinary Medicine, UC Davis, Davis, CA 95616, USA
| | | | | |
Collapse
|
43
|
Dockray GJ, Burdyga G. Plasticity in vagal afferent neurones during feeding and fasting: mechanisms and significance. Acta Physiol (Oxf) 2011; 201:313-21. [PMID: 21062423 DOI: 10.1111/j.1748-1716.2010.02219.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The ingestion of food activates mechanisms leading to inhibition of food intake and gastric emptying mediated by the release of regulatory peptides, for example cholecystokinin (CCK), and lipid amides, e.g. oleylethanolamide from the gut. In addition, there are both peptides (e.g. ghrelin) and lipid amides (e.g. anandamide) that appear to signal the absence of food in the gut and that are associated with the stimulation of food intake. Vagal afferent neurones are a common target for both types of signal. Remarkably, the neurochemical phenotype of these neurones itself depends on nutritional status. CCK acting at CCK1 receptors on vagal afferent neurones stimulates expression in these neurones of Y2-receptors and the neuropeptide CART, both of which are associated with the inhibition of food intake. Conversely, in fasted rats when plasma CCK is low, these neurones express cannabinoid (CB)-1 and melanin concentrating hormone (MCH)-1 receptors, and MCH, and this is inhibited by exogenous CCK or endogenous CCK released by refeeding. The stimulation of CART expression by CCK is mediated by the activation of CREB and EGR1; ghrelin inhibits the action of CCK by promoting nuclear exclusion of CREB and leptin potentiates the action of CCK by the stimulation of EGR1 expression. Vagal afferent neurones therefore constitute a level of integration outside the CNS for nutrient-derived signals that control energy intake and that are capable of encoding recent nutrient ingestion.
Collapse
Affiliation(s)
- G J Dockray
- Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, Crown Street, Liverpool, UK.
| | | |
Collapse
|
44
|
Current world literature. Curr Opin Endocrinol Diabetes Obes 2011; 18:83-98. [PMID: 21178692 DOI: 10.1097/med.0b013e3283432fa7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
45
|
Lee J, Martin E, Paulino G, de Lartigue G, Raybould HE. Effect of ghrelin receptor antagonist on meal patterns in cholecystokinin type 1 receptor null mice. Physiol Behav 2011; 103:181-7. [PMID: 21277881 DOI: 10.1016/j.physbeh.2011.01.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Revised: 01/07/2011] [Accepted: 01/19/2011] [Indexed: 11/24/2022]
Abstract
Vagal afferent neurons (VAN) express the cholecystokinin (CCK) type 1 receptor (CCK₁R) and, as predicted by the role of CCK in inducing satiation, CCK₁R⁻/⁻ mice ingest larger and longer meals. However, after a short fast, CCK₁R⁻/⁻ mice ingesting high fat (HF) diets initiate feeding earlier than wild-type mice. We hypothesized that the increased drive to eat in CCK₁R⁻/⁻ mice eating HF diet is mediated by ghrelin, a gut peptide that stimulates food intake. The decrease in time to first meal, and the increase in meal size and duration in CCK₁R⁻/⁻ compared to wild-type mice ingesting high fat (HF) diet were reversed by administration of GHSR1a antagonist D-(Lys3)-GHRP-6 (p<0.05). Administration of the GHSR1a antagonist significantly increased expression of the neuropeptide cocaine and amphetamine-regulated transcript (CART) in VAN of HF-fed CCK₁R⁻/⁻ but not wild-type mice. Administration of the GHSR1a antagonist decreased neuronal activity measured by immunoreactivity for fos protein in the nucleus of the solitary tract (NTS) and the arcuate nucleus of both HF-fed wild-type and CCK₁R⁻/⁻ mice. The data show that hyperphagia in CCK₁R⁻/⁻ mice ingesting HF diet is reversed by blockade of the ghrelin receptor, suggesting that in the absence of the CCK₁R, there is an increased ghrelin-dependent drive to feed. The site of action of ghrelin receptors is unclear, but may involve an increase in expression of CART peptide in VAN in HF-fed CCK₁R⁻/⁻ mice.
Collapse
Affiliation(s)
- Jennifer Lee
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, UC Davis, Davis, CA 95616, USA
| | | | | | | | | |
Collapse
|
46
|
de Lartigue G, Lur G, Dimaline R, Varro A, Raybould H, Dockray GJ. EGR1 Is a target for cooperative interactions between cholecystokinin and leptin, and inhibition by ghrelin, in vagal afferent neurons. Endocrinology 2010; 151:3589-99. [PMID: 20534729 PMCID: PMC2940532 DOI: 10.1210/en.2010-0106] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Food intake is regulated by signals from peripheral organs, but the way these are integrated remains uncertain. Cholecystokinin (CCK) from the intestine and leptin from adipocytes interact to inhibit food intake. Our aim was to examine the hypothesis that these interactions occur at the level of vagal afferent neurons via control of the immediate early gene EGR1. We now report that CCK stimulates redistribution to the nucleus of early growth response factor-1 (EGR1) in these neurons in vivo and in culture, and these effects are not dependent on EGR1 synthesis. Leptin stimulates EGR1 expression; leptin alone does not stimulate nuclear translocation, but it strongly potentiates the action of CCK. Ghrelin inhibits CCK-stimulated nuclear translocation of EGR1 and leptin-stimulated EGR1 expression. Expression of the gene encoding the satiety peptide cocaine- and amphetamine-regulated transcript (CARTp) is stimulated by CCK via an EGR1-dependent mechanism, and this is strongly potentiated by leptin. Leptin potentiated inhibition of food intake by endogenous CCK in the rat in conditions reflecting changes in EGR1 activation. The data indicate that by separately regulating EGR1 activation and synthesis, CCK and leptin interact cooperatively to define the capacity for satiety signaling by vagal afferent neurons; manipulation of these interactions may be therapeutically beneficial.
Collapse
Affiliation(s)
- Guillaume de Lartigue
- Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, Liverpool, UK.
| | | | | | | | | | | |
Collapse
|
47
|
Burdyga G, Varro A, Dimaline R, Thompson DG, Dockray GJ. Expression of cannabinoid CB1 receptors by vagal afferent neurons: kinetics and role in influencing neurochemical phenotype. Am J Physiol Gastrointest Liver Physiol 2010; 299:G63-9. [PMID: 20430875 PMCID: PMC2904113 DOI: 10.1152/ajpgi.00059.2010] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The intestinal hormone cholecystokinin (CCK) inhibits food intake via stimulation of vagal afferent neurons (VAN). Recent studies suggest that CCK also regulates the expression of some G protein-coupled receptors and neuropeptide transmitters in these neurons. The aim of the present study was to characterize the expression of cannabinoid (CB)1 receptors in VAN and to determine whether stimulation of these receptors plays a role in regulating neurochemical phenotype. Expression of CB1 in rat VAN was detectable by in situ hybridization or immunohistochemistry after 6 h of fasting and increased to a maximum after 24 h when approximately 50% of neurons in the mid and caudal regions expressed the receptor. Melanin-concentrating hormone (MCH)1 receptors also increased with fasting, but the changes were delayed compared with CB1; in contrast Y2 receptors (Y2R) exhibited reciprocal changes in expression to CB1. Administration of CCK8s (10 nmol ip) to fasted rats decreased expression of CB1 with a t(1/2) of approximately 1 h compared with 3 h for MCH1. The action of CCK8s was inhibited by ghrelin and orexin-A. The CB1 agonist anandamide (intraperitoneally) reversed the effect of CCK8s on CB1, MCH1, and Y2 receptor expression. In contrast, in rats fasted for 18 h, administration of a CB1 antagonist/inverse agonist (AM281 ip) downregulated CB1 expression and increased Y2 receptor expression. Activation of vagal CB1 receptors therefore influences the neurochemical phenotype of these neurons, indicating a new and hitherto unrecognized role for endocannabinoids in gut-brain signaling.
Collapse
Affiliation(s)
- Galina Burdyga
- 1Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, Liverpool;
| | - Andrea Varro
- 1Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, Liverpool;
| | - Rod Dimaline
- 1Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, Liverpool;
| | - David G. Thompson
- 2Division of Gastroenterology, Hope University Hospital, Salford, United Kingdom
| | - Graham J. Dockray
- 1Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, Liverpool;
| |
Collapse
|