1
|
Chen S, Bi Y, Zhang L. ASS1 is a hub gene and possible therapeutic target for regulating metabolic dysfunction-associated steatotic liver disease modulated by a carbohydrate-restricted diet. Mol Divers 2025:10.1007/s11030-025-11187-6. [PMID: 40244373 DOI: 10.1007/s11030-025-11187-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the leading cause of chronic liver disease globally. A low-carbohydrate diet (LCD) offers benefits to MASLD patients, albeit its exact mechanism is not fully understood. Using public liver transcriptome data from MASLD patients before/after LCD intervention, we applied differential expression analysis and machine learning to identify key genes. We initially identified 162 differentially expressed genes in the GSE107650 dataset. Secondly, employing two machine learning algorithms, we found that PRAMENP, LEAP2, LOC105379013, and argininosuccinate synthetase 1 (ASS1) are potential hub genes. Additionally, protein-protein interaction and single-cell RNA location analyses suggested that ASS1 was the most crucial hub gene. Then, L1000CDS2 analysis of the gene-expression signatures was employed for drug repurposing studies. CGP71683, an appetite suppressant, was predicted to improve MASLD and may mimic the ASS1 expression pattern induced by an LCD. Molecular dynamics confirmed spontaneous, stable CGP71683-ASS1 complex formation. Overall, this work based on analysis of machine learning algorithms, essential gene identification, and drug repurposing studies suggested that ASS1 is an essential gene in MASLD and CGP71683 is a potential drug candidate for treating MASLD by targeting ASS1 and mimicking the beneficial effects of an LCD. However, due to the inherent limitations of a purely computational approach, further experimental investigation is necessary to validate the anticipated results.
Collapse
Affiliation(s)
- Shaojun Chen
- College of Traditional Chinese Medicine, Zhejiang Pharmaceutical University, Ningbo, 315000, China
| | - Yanhua Bi
- The Children's Hospital, National Clinical Research Centre for Child Health, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Lihua Zhang
- College of Food Science and Technology, Zhejiang Pharmaceutical University, Ningbo, 315000, China.
| |
Collapse
|
2
|
Huang YJ, Cao ZF, Wang J, Yang J, Wei YJ, Tang YC, Cheng YX, Zhou J, Zhang ZX. Why MUC16 mutations lead to a better prognosis: A study based on The Cancer Genome Atlas gastric cancer cohort. World J Clin Cases 2021; 9:4143-4158. [PMID: 34141777 PMCID: PMC8173414 DOI: 10.12998/wjcc.v9.i17.4143] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/25/2021] [Accepted: 03/05/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND MUC16, encoding cancer antigen 125, is a frequently mutated gene in gastric cancer. In addition, MUC16 mutations seem to result in a better prognosis in gastric cancer. However, the mechanisms that lead to a better prognosis by MUC16 mutations have not yet been clarified. AIM To delve deeper into the underlying mechanisms that explain why MUC16 mutations signal a better prognosis in gastric cancer. METHODS We used multi-omics data, including mRNA, simple nucleotide variation, copy number variation and methylation data from The Cancer Genome Atlas, to explore the relationship between MUC16 mutations and prognosis. Cox regression and random survival forest algorithms were applied to search for hub genes. Gene set enrichment analysis was used to elucidate the molecular mechanisms. Single-sample gene set enrichment analysis and "EpiDISH" were used to assess immune cells infiltration, and "ESTIMATE" for analysis of the tumor microenvironment. RESULTS Our study found that compared to the wild-type group, the mutation group had a better prognosis. Additional analysis indicated that the MUC16 mutations appear to activate the DNA repair and p53 pathways to act as an anti-tumor agent. We also identified a key gene, NPY1R (neuropeptide Y receptor Y1), which was significantly more highly expressed in the MUC16 mutations group than in the MUC16 wild-type group. The high expression of NPY1R predicted a poorer prognosis, which was also confirmed in a separate Gene Expression Omnibus cohort. Further susceptibility analysis revealed that NPY1R might be a potential drug target for gastric cancer. Furthermore, in the analysis of the tumor microenvironment, we found that immune cells in the mutation group exhibited higher anti-tumor effects. In addition, the tumor mutation burden and cancer stem cells index were also higher in the mutation group than in the wild-type group. CONCLUSION We speculated that the MUC16 mutations might activate the p53 pathway and DNA repair pathway: alternatively, the tumor microenvironment may be involved.
Collapse
Affiliation(s)
- Yu-Jie Huang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Zhi-Fei Cao
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu Province, China
| | - Jie Wang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Jian Yang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Yi-Jun Wei
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Yu-Chen Tang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Yin-Xiang Cheng
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Jian Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Zi-Xiang Zhang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| |
Collapse
|
3
|
Richardson RR, Groenen M, Liu M, Mountford SJ, Briddon SJ, Holliday ND, Thompson PE. Heterodimeric Analogues of the Potent Y1R Antagonist 1229U91, Lacking One of the Pharmacophoric C-Terminal Structures, Retain Potent Y1R Affinity and Show Improved Selectivity over Y4R. J Med Chem 2020; 63:5274-5286. [PMID: 32364733 DOI: 10.1021/acs.jmedchem.0c00027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The cyclic dimeric peptide 1229U91 (GR231118) has an unusual structure and displays potent, insurmountable antagonism of the Y1 receptor. To probe the structural basis for this activity, we have prepared ring size variants and heterodimeric compounds, identifying the specific residues underpinning the mechanism of 1229U91 binding. The homodimeric structure was shown to be dispensible, with analogues lacking key pharmacophoric residues in one dimer arm retaining high antagonist affinity. Compounds 11d-h also showed enhanced Y1R selectivity over Y4R compared to 1229U91.
Collapse
Affiliation(s)
- Rachel R Richardson
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Parkville, Victoria 3052, Australia.,Institute of Cell Signalling, School of Biomedical Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, U.K
| | - Marleen Groenen
- Institute of Cell Signalling, School of Biomedical Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, U.K
| | - Mengjie Liu
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Simon J Mountford
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Stephen J Briddon
- Institute of Cell Signalling, School of Biomedical Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, U.K
| | - Nicholas D Holliday
- Institute of Cell Signalling, School of Biomedical Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, U.K
| | - Philip E Thompson
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Parkville, Victoria 3052, Australia
| |
Collapse
|
4
|
Distribution of D1 and D2 receptor- immunoreactive neurons in the paraventricular nucleus of the hypothalamus in the rat. J Chem Neuroanat 2019; 98:97-103. [DOI: 10.1016/j.jchemneu.2019.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 04/08/2019] [Accepted: 04/08/2019] [Indexed: 12/12/2022]
|
5
|
Wang C, Han X, Sun X, Guo F, Luan X, Xu L. Orexin-A signaling in the paraventricular nucleus promote gastric acid secretion and gastric motility through the activation neuropeptide Y Y 1 receptors and modulated by the hypothalamic lateral area. Neuropeptides 2019; 74:24-33. [PMID: 30700376 DOI: 10.1016/j.npep.2019.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 01/22/2019] [Accepted: 01/22/2019] [Indexed: 01/23/2023]
Abstract
OBJECTIVE Abnormal gastric acid secretion and gastric dyskinesia are common gastroenterological ailments. Our study aims to investigate the effect of orexin-A in the paraventricular nucleus (PVN) gastric motility and gastric acid secretion. METHODS The source of orexin-A neuronal projections to the PVN were explored by retrograde tracing and fluorescence immunohistochemistry experiments. Neuronal discharge recordings of single cells were taken within the PVN. Gastric motility was recorded using a force transducer implanted into the stomach, and gastric acid secretion measured through a pyloric catheter. RESULTS Orexin-A-positive neuronal projections from LHA to PVN were found. Administration of orexin-A to PVN activated the firing of 63.2% NPY-excited/GD-excitatory (GD-E) neurons but suppressed the firing of 55.9% NPY-inhibited/GD-inhibitory (GD-I) neurons, promoted gastric motility and gastric acid secretion in a dose-dependent manner. Responses produced by orexin-A could be partially blocked by Y1 receptor antagonist GR-231118; Electrical stimulation to the the hypothalamic lateral area (LHA) altered NPY-sensitive/GD neuronal activity in the PVN, stimulated gastric motility and gastric acid secretion. Additionally, these effects induced by LHA electrical stimulation were blocked by administration of the OX1R antagonist SB-334867 to the PVN. CONCLUSION Orexin-A from LHA neurons act on the PVN to enhance gastric motility and gastric acid secretion, with Y1 receptor signaling playing a critical role.
Collapse
Affiliation(s)
- Cheng Wang
- Qingdao University, School of Basic Medical Sciences, Shandong, Qingdao 266071, China
| | - Xiaohua Han
- Qingdao University, School of Basic Medical Sciences, Shandong, Qingdao 266071, China
| | - Xiangrong Sun
- Qingdao University, School of Basic Medical Sciences, Shandong, Qingdao 266071, China
| | - Feiei Guo
- Qingdao University, School of Basic Medical Sciences, Shandong, Qingdao 266071, China
| | - Xiao Luan
- Qingdao University, School of Basic Medical Sciences, Shandong, Qingdao 266071, China
| | - Luo Xu
- Qingdao University, School of Basic Medical Sciences, Shandong, Qingdao 266071, China.
| |
Collapse
|
6
|
Shareghi Brojeni M, Salimi M, Mirmohammadsadeghi Z, Haghparast A, Eliassi A. Comparison of Effects of Light Anesthetics, Diethyl Ether and Carbon Dioxide, on Hypothalamic Paraventricular Nucleus D 1 and D 2 Dopamine Receptors- and Glucosensitive Neurons-Induced Food Intake in Fasted Conscious Rats. Basic Clin Neurosci 2018; 9:269-274. [PMID: 30519385 PMCID: PMC6276533 DOI: 10.32598/bcn.9.4.269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 05/15/2017] [Accepted: 07/30/2017] [Indexed: 11/20/2022] Open
Abstract
Introduction Carbon Dioxide (CO2) and diethyl ether are used as light anesthetics. However, experimental data about their side effects are scarce. In addition, in all our previous works on regulatory mechanisms of hypothalamus during food intake, including the effect of Paraventricular Nucleus (PVN) D1 and D2 dopamine receptors and glucosensitive neurons, the drug injections were performed under brief diethyl ether anesthesia. In the current study, we tested the hypothesis which postulates that CO2 and diethyl ether as light anesthetic agents affect the stimulatory effect of PVN dopamine receptors and glucosensitive neurons in feeding behavior. Methods Male Wistar rats were implanted with guide cannula directed to their PVN. Glucose (0.8 μg), SKF38393 (D1 agonist, 0.5 μg), quinpirole (D2 agonist, 0.3 μg) and saline (0.3 μL) were microinjected into the PVN and food intake was measured over 1 hour. Results Our results showed that CO2 but not diethyl ether decreased food intake compared to intact animals. The PVN injections of glucose, SKF38393, and quinpirole increased food intake under brief diethyl ether anesthesia. In contrast, the PVN microinjected glucose-induced and dopamine receptor agonists-induced food intake were inhibited under light CO2 anesthesia. Conclusion Our results suggest that brief exposure to CO2 and diethyl ether as light anesthetic agents may affect PVN glucosensing neurons-induced and dopamine receptors-induced food intake in fasted rats.
Collapse
Affiliation(s)
- Masoud Shareghi Brojeni
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Morteza Salimi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Mirmohammadsadeghi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afsaneh Eliassi
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Whole genome MBD-seq and RRBS analyses reveal that hypermethylation of gastrointestinal hormone receptors is associated with gastric carcinogenesis. Exp Mol Med 2018; 50:1-14. [PMID: 30510283 PMCID: PMC6277407 DOI: 10.1038/s12276-018-0179-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 07/16/2018] [Accepted: 08/09/2018] [Indexed: 12/15/2022] Open
Abstract
DNA methylation is a regulatory mechanism in epigenetics that is frequently altered during human carcinogenesis. To detect critical methylation events associated with gastric cancer (GC), we compared three DNA methylomes from gastric mucosa (GM), intestinal metaplasia (IM), and gastric tumor (GT) cells that were microscopically dissected from an intestinal-type early gastric cancer (EGC) using methylated DNA binding domain sequencing (MBD-seq) and reduced representation bisulfite sequencing (RRBS) analysis. In this study, we focused on differentially methylated promoters (DMPs) that could be directly associated with gene expression. We detected 2,761 and 677 DMPs between the GT and GM by MBD-seq and RRBS, respectively, and for a total of 3,035 DMPs. Then, 514 (17%) of all DMPs were detected in the IM genome, which is a precancer of GC, supporting that some DMPs might represent an early event in gastric carcinogenesis. A pathway analysis of all DMPs demonstrated that 59 G protein-coupled receptor (GPCR) genes linked to the hypermethylated DMPs were significantly enriched in a neuroactive ligand–receptor interaction pathway. Furthermore, among the 59 GPCRs, six GI hormone receptor genes (NPY1R, PPYR1, PTGDR, PTGER2, PTGER3, and SSTR2) that play an inhibitory role in the secretion of gastrin or gastric acid were selected and validated as potential biomarkers for the diagnosis or prognosis of GC patients in two cohorts. These data suggest that the loss of function of gastrointestinal (GI) hormone receptors by promoter methylation may lead to gastric carcinogenesis because gastrin and gastric acid have been known to play a role in cell differentiation and carcinogenesis in the GI tract. A sequencing study reveals abnormal changes to DNA that set the stage for stomach cancer development. DNA methylation, the addition of methyl groups to alter DNA activity, is often disrupted in human cancers. Yong Sung Kim at the Korea Research Institute of Bioscience and Biotechnology (KRIBB) in Daejeon, South Korea, and co-workers used sequencing technogy to identify critical methylation changes in stomach epithelial cells, intestinal metaplasia lesion and tumor cells during early-stage gastric cancer. The team found 3,035 abnormally methylated DNA regions related to the expression of particular genes. Further analysis identified six hormone receptor genes directly involved with stomach acid secretion, whose altered expression was linked to over-methylated DNA regions. Loss of function within these six genes may lead to gastric cancer, and their expression levels could be valuable biomarkers for the disease.
Collapse
|
8
|
Kermani M, Fatahi Z, Sun D, Haghparast A, French C. Operant Protocols for Assessing the Cost-benefit Analysis During Reinforced Decision Making by Rodents. J Vis Exp 2018. [PMID: 30247477 DOI: 10.3791/57907] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Reinforcement-guided decision making is the ability to choose between competing courses of action based on the relative value of the benefits and their consequences. This process is integral to the normal human behavior and has been shown to be disrupted by neurological and psychiatric disorders such as addiction, schizophrenia, and depression. Rodents have long been used to uncover the neurobiology of human cognition. To this end, several behavioral tasks have been developed; however, most are non-automated and are labor-intensive. The recent development of the open-source microcontroller has enabled researchers to automate operant-based tasks for assessing a variety of cognitive tasks, standardizing the stimulus presentation, improving the data recording and consequently, improving the research output. Here, we describe an automated delay-based reinforcement-guided decision-making task, using an operant T-maze controlled by custom-written software programs. Using these decision-making tasks, we show the changes in the local field potential activities in the anterior cingulate cortex of a rat whilst it performs a delay-based cost-and-benefit decision-making task.
Collapse
Affiliation(s)
- Mojtaba Kermani
- Department of Optometry and Vision Science, The University of Melbourne; Department of Medicine, The University of Melbourne;
| | - Zahra Fatahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Science
| | - Dechuan Sun
- Department of Medicine, The University of Melbourne
| | - Abbas Haghparast
- Neuroscience Research Center, Shahid Beheshti University of Medical Science
| | - Chris French
- Department of Medicine, The University of Melbourne; Royal Melbourne Hospital
| |
Collapse
|
9
|
Luan X, Sun X, Guo F, Zhang D, Wang C, Ma L, Xu L. Lateral hypothalamic Orexin-A-ergic projections to the arcuate nucleus modulate gastric functionin vivo. J Neurochem 2017; 143:697-707. [PMID: 28984906 DOI: 10.1111/jnc.14233] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 09/18/2017] [Accepted: 09/22/2017] [Indexed: 02/06/2023]
Affiliation(s)
- Xiao Luan
- Department of Pathophysiology; Medical College of Qingdao University; Qingdao China
| | - Xiangrong Sun
- Department of Pathophysiology; Medical College of Qingdao University; Qingdao China
| | - Feifei Guo
- Department of Pathophysiology; Medical College of Qingdao University; Qingdao China
| | - Di Zhang
- Department of Pathophysiology; Medical College of Qingdao University; Qingdao China
| | - Cheng Wang
- Department of Pathophysiology; Medical College of Qingdao University; Qingdao China
| | - Li Ma
- Departmemt of Clinical Nutrition; Affiliated Hospital; Qingdao University; Qingdao China
| | - Luo Xu
- Department of Pathophysiology; Medical College of Qingdao University; Qingdao China
| |
Collapse
|
10
|
Mirmohammadsadeghi Z, Shareghi Brojeni M, Haghparast A, Eliassi A. Role of paraventricular hypothalamic dopaminergic D 1 receptors in food intake regulation of food-deprived rats. Eur J Pharmacol 2017; 818:43-49. [PMID: 29056523 DOI: 10.1016/j.ejphar.2017.10.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 10/15/2017] [Accepted: 10/17/2017] [Indexed: 12/28/2022]
Abstract
Dopaminergic neurons play an important role on central regulatory mechanisms of feeding behavior. Dopamine receptors are distributed within the hypothalamus and densely localized in the paraventricular hypothalamic nucleus (PVN). From these ideas we postulated that PVN D1 receptors may play a role in regulating the food intake behavioral process. In this paper, we considered the effects of SKF38393, a D1 receptor agonist, and the D1 receptor antagonist (SCH23390), on food intake of conscious rats deprived of food for 24h. Our findings revealed that intraparaventricular injections of SKF383993 (0.3-5µg) stimulated food intake behavior in a dose dependent manner. This stimulatory effect of SKF3833 persisted over 2h of the monitoring period. The PVN injections of D1 receptor antagonist were associated with dose-dependent inhibition of food intake. SCH23390 (0.01µg) was also administered 5min before intraparaventricular injection of SKF3833. The results showed that SCH23390 suppressed stimulated food intake induced by SKF38393 (1.2µg). In conclusion, endogenous dopamine impact PVN D1 receptors and may be a factor in regulating the food intake behavioral process.
Collapse
Affiliation(s)
| | - Masoud Shareghi Brojeni
- Department of Physiology, Medical School, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afsaneh Eliassi
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Physiology, Medical School, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Sun S, Xu L, Sun X, Guo F, Gong Y, Gao S. Orexin-A affects gastric distention sensitive neurons in the hippocampus and gastric motility and regulation by the perifornical area in rats. Neurosci Res 2016; 110:59-67. [PMID: 27080329 DOI: 10.1016/j.neures.2016.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 03/25/2016] [Accepted: 04/01/2016] [Indexed: 12/20/2022]
Abstract
Orexin-A is mainly produced in the lateral hypothalamus (LHA) and the perifornical area (PeF). Here, we aim to elucidate the effects of orexin-A in the hippocampus (Hi) on gastric distention (GD)-sensitive neurons and gastric motility, and potential regulation mechanisms by the PeF. Retrograde tracing and fluorescent-immunohistochemical staining were used to determine orexin-A neuronal projections. Single unit discharges in the Hi were recorded extracellularly and gastric motility in conscious rats was monitored during administration of orexin-A to the Hi or electrical stimulation of the PeF. Orexin-A administration to the Hi excited most of the GD-excitatory (GD-E) neurons and GD-inhibitory (GD-I) neurons, and increased gastric motility in a dose-dependent manner. All of effects induced by orexin-A could be partly blocked by pretreatment with orexin-A antagonist, SB-334867. Electrical stimulation of the PeF excited the majority of the orexin-A-responsive GD neurons in the Hi and promoted gastric motility. Additionally, pretreatment with SB-334867 in the Hi increased the firing rate of GDI and GDE neurons following electrical stimulation of the PeF. These findings suggest that orexin-A could regulate activities of GD-sensitive neurons and gastric motility. Furthermore, the PeF may be involved in this regulatory pathway.
Collapse
Affiliation(s)
- Shu Sun
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao 266021, China
| | - Luo Xu
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao 266021, China.
| | - Xiangrong Sun
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao 266021, China
| | - Feifei Guo
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao 266021, China
| | - Yanling Gong
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Shengli Gao
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao 266021, China
| |
Collapse
|
12
|
Abstract
AgRP and POMC neurons are two key cell types that regulate feeding in response to hormones and nutrients. Recently, it was discovered that these neurons are also rapidly modulated by the mere sight and smell of food. This rapid sensory regulation "resets" the activity of AgRP and POMC neurons before a single bite of food has been consumed. This surprising and counterintuitive discovery challenges longstanding assumptions about the function and regulation of these cells. Here we review these recent findings and discuss their implications for our understanding of feeding behavior. We propose several alternative hypotheses for how these new observations might be integrated into a revised model of the feeding circuit, and also highlight some of the key questions that remain to be answered.
Collapse
Affiliation(s)
- Yiming Chen
- Department of Physiology, University of California, San Francisco, CA, USA
| | - Zachary A Knight
- Department of Physiology, University of California, San Francisco, CA, USA
| |
Collapse
|
13
|
Li J, Hu Z, de Lecea L. The hypocretins/orexins: integrators of multiple physiological functions. Br J Pharmacol 2014; 171:332-50. [PMID: 24102345 DOI: 10.1111/bph.12415] [Citation(s) in RCA: 186] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Revised: 07/16/2013] [Accepted: 08/02/2013] [Indexed: 12/28/2022] Open
Abstract
The hypocretins (Hcrts), also known as orexins, are two peptides derived from a single precursor produced in the posterior lateral hypothalamus. Over the past decade, the orexin system has been associated with numerous physiological functions, including sleep/arousal, energy homeostasis, endocrine, visceral functions and pathological states, such as narcolepsy and drug abuse. Here, we review the discovery of Hcrt/orexins and their receptors and propose a hypothesis as to how the orexin system orchestrates these multifaceted physiological functions.
Collapse
Affiliation(s)
- Jingcheng Li
- Department of Physiology, Third Military Medical University, Chongqing, China
| | | | | |
Collapse
|