1
|
Tanday N, Zhu W, Tarasov AI, Flatt PR, Irwin N. [P 3]PP, a stable, long-acting pancreatic polypeptide analogue, evokes weight lowering and pancreatic beta-cell-protective effects in obesity-associated diabetes. Diabetes Obes Metab 2024; 26:4945-4957. [PMID: 39192525 DOI: 10.1111/dom.15897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 08/29/2024]
Abstract
AIM To thoroughly investigate the impact of sustained neuropeptide Y4 receptor (NPY4R) activation in obesity-associated diabetes. METHODS Initially, the prolonged pharmacodynamic profile of the enzymatically stable pancreatic polypeptide (PP) analogue, [P3]PP, was confirmed in normal mice up to 24 h after injection. Subsequent to this, [P3]PP was administered twice daily (25 nmol/kg) for 28 days to high-fat-fed mice with streptozotocin-induced insulin deficiency, known as HFF/STZ mice. RESULTS Treatment with [P3]PP for 28 days reduced energy intake and was associated with notable weight loss. In addition, circulating glucose was returned to values of approximately 8 mmol/L in [P3]PP-treated mice, with significantly increased plasma insulin and decreased glucagon concentrations. Glucose tolerance and glucose-stimulated insulin secretion were improved in [P3]PP-treated HFF/STZ mice, with no obvious effect on peripheral insulin sensitivity. Benefits on insulin secretion were associated with elevated pancreatic insulin content as well as islet and beta-cell areas. Positive effects on islet architecture were linked to increased beta-cell proliferation and decreased apoptosis. Treatment intervention also decreased islet alpha-cell area, but pancreatic glucagon content remained unaffected. In addition, [P3]PP-treated HFF/STZ mice presented with reduced plasma alanine transaminase and aspartate transaminase levels, with no change in circulating amylase concentrations. In terms of plasma lipid profile, triglyceride and cholesterol levels were significantly decreased by [P3]PP treatment, when compared to saline controls. CONCLUSION Collectively, these data highlight for the first time the potential of enzymatically stable PP analogues for the treatment of obesity and related diabetes.
Collapse
Affiliation(s)
- Neil Tanday
- Diabetes Research Centre, Ulster University, Coleraine, UK
| | - Wuyun Zhu
- Diabetes Research Centre, Ulster University, Coleraine, UK
| | | | - Peter R Flatt
- Diabetes Research Centre, Ulster University, Coleraine, UK
| | - Nigel Irwin
- Diabetes Research Centre, Ulster University, Coleraine, UK
| |
Collapse
|
2
|
Lafferty RA, Flatt PR, Irwin N. NPYR modulation: Potential for the next major advance in obesity and type 2 diabetes management? Peptides 2024; 179:171256. [PMID: 38825012 DOI: 10.1016/j.peptides.2024.171256] [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: 03/15/2024] [Revised: 05/13/2024] [Accepted: 05/30/2024] [Indexed: 06/04/2024]
Abstract
The approval of the glucagon-like peptide 1 (GLP-1) mimetics semaglutide and liraglutide for management of obesity, independent of type 2 diabetes (T2DM), has initiated a resurgence of interest in gut-hormone derived peptide therapies for the management of metabolic diseases, but side-effect profile is a concern for these medicines. However, the recent approval of tirzepatide for obesity and T2DM, a glucose-dependent insulinotropic polypeptide (GIP), GLP-1 receptor co-agonist peptide therapy, may provide a somewhat more tolerable option. Despite this, an increasing number of non-incretin alternative peptides are in development for obesity, and it stands to reason that other hormones will take to the limelight in the coming years, such as peptides from the neuropeptide Y family. This narrative review outlines the therapeutic promise of the neuropeptide Y family of peptides, comprising of the 36 amino acid polypeptides neuropeptide Y (NPY), peptide tyrosine-tyrosine (PYY) and pancreatic polypeptide (PP), as well as their derivatives. This family of peptides exerts a number of metabolically relevant effects such as appetite regulation and can influence pancreatic beta-cell survival. Although some of these actions still require full translation to the human setting, potential therapeutic application in obesity and type 2 diabetes is conceivable. However, like GLP-1 and GIP, the endogenous NPY, PYY and PP peptide forms are subject to rapid in vivo degradation and inactivation by the serine peptidase, dipeptidyl-peptidase 4 (DPP-4), and hence require structural modification to prolong circulating half-life. Numerous protective modification strategies are discussed in this regard herein, alongside related impact on biological activity profile and therapeutic promise.
Collapse
Affiliation(s)
- Ryan A Lafferty
- Diabetes Research Centre, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK.
| | - Peter R Flatt
- Diabetes Research Centre, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK
| | - Nigel Irwin
- Diabetes Research Centre, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK
| |
Collapse
|
3
|
Reed J, Bain SC, Kanamarlapudi V. The Regulation of Metabolic Homeostasis by Incretins and the Metabolic Hormones Produced by Pancreatic Islets. Diabetes Metab Syndr Obes 2024; 17:2419-2456. [PMID: 38894706 PMCID: PMC11184168 DOI: 10.2147/dmso.s415934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/07/2024] [Indexed: 06/21/2024] Open
Abstract
In healthy humans, the complex biochemical interplay between organs maintains metabolic homeostasis and pathological alterations in this process result in impaired metabolic homeostasis, causing metabolic diseases such as diabetes and obesity, which are major global healthcare burdens. The great advancements made during the last century in understanding both metabolic disease phenotypes and the regulation of metabolic homeostasis in healthy individuals have yielded new therapeutic options for diseases like type 2 diabetes (T2D). However, it is unlikely that highly desirable more efficacious treatments will be developed for metabolic disorders until the complex systemic regulation of metabolic homeostasis becomes more intricately understood. Hormones produced by pancreatic islet beta-cells (insulin) and alpha-cells (glucagon) are pivotal for maintaining metabolic homeostasis; the activity of insulin and glucagon are reciprocally correlated to achieve strict control of glucose levels (normoglycaemia). Metabolic hormones produced by other pancreatic islet cells and incretins produced by the gut are also crucial for maintaining metabolic homeostasis. Recent studies highlighted the incomplete understanding of metabolic hormonal synergism and, therefore, further elucidation of this will likely lead to more efficacious treatments for diseases such as T2D. The objective of this review is to summarise the systemic actions of the incretins and the metabolic hormones produced by the pancreatic islets and their interactions with their respective receptors.
Collapse
Affiliation(s)
- Joshua Reed
- Institute of Life Science, Medical School, Swansea University, Swansea, SA2 8PP, UK
| | - Stephen C Bain
- Institute of Life Science, Medical School, Swansea University, Swansea, SA2 8PP, UK
| | | |
Collapse
|
4
|
Zhang L, Herzog H. Important role of NPY-Y4R signalling in the dual control of feeding and physical activity. Neuropeptides 2024; 105:102425. [PMID: 38554699 DOI: 10.1016/j.npep.2024.102425] [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: 01/17/2024] [Revised: 03/14/2024] [Accepted: 03/16/2024] [Indexed: 04/02/2024]
Abstract
The control of feeding and physical activity is tightly linked and coordinated. However the underlying mechanisms are unclear. One of the major regulatory systems of feeding behaviour involves neuropeptide Y (NPY) signalling, with the signalling mediated through NPY Y4 receptor also known to influence activity. Here we show that mice globally lacking the Npy4r (Npy4r-/-) in the absence of access to a running wheel behaved WT-like with regards to food intake, energy expenditure, respiratory exchange ratio and locomotion regardless of being fed on a chow or high fat diet. Interestingly however, when given the access to a running wheel, Npy4r-/- mice while having a comparable locomotor activity, showed significantly higher wheel-running activity than WT, again regardless of dietary conditions. This higher wheel-running activity in Npy4r-/-mice arose from an increased dark-phase running time rather than changes in number of running bouts or the running speed. Consistently, energy expenditure was higher in Npy4r-/- than WT mice. Importantly, food intake was reduced in Npy4r-/-mice under wheel access condition which was due to decreased feeding bouts rather than changes in meal size. Together, these findings demonstrate an important role of Npy4r signalling in the dual control of feeding and physical activity, particularly in the form of wheel-running activity.
Collapse
Affiliation(s)
- Lei Zhang
- St Vincent's Centre for Applied Medical Research, School of Clinical Medicine, UNSW Medicine and Health, UNSW SYDNEY, NSW 2052, Australia.
| | - Herbert Herzog
- St Vincent's Centre for Applied Medical Research, School of Clinical Medicine, UNSW Medicine and Health, UNSW SYDNEY, NSW 2052, Australia
| |
Collapse
|
5
|
Gan HW, Cerbone M, Dattani MT. Appetite- and Weight-Regulating Neuroendocrine Circuitry in Hypothalamic Obesity. Endocr Rev 2024; 45:309-342. [PMID: 38019584 PMCID: PMC11074800 DOI: 10.1210/endrev/bnad033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 10/25/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023]
Abstract
Since hypothalamic obesity (HyOb) was first described over 120 years ago by Joseph Babinski and Alfred Fröhlich, advances in molecular genetic laboratory techniques have allowed us to elucidate various components of the intricate neurocircuitry governing appetite and weight regulation connecting the hypothalamus, pituitary gland, brainstem, adipose tissue, pancreas, and gastrointestinal tract. On a background of an increasing prevalence of population-level common obesity, the number of survivors of congenital (eg, septo-optic dysplasia, Prader-Willi syndrome) and acquired (eg, central nervous system tumors) hypothalamic disorders is increasing, thanks to earlier diagnosis and management as well as better oncological therapies. Although to date the discovery of several appetite-regulating peptides has led to the development of a range of targeted molecular therapies for monogenic obesity syndromes, outside of these disorders these discoveries have not translated into the development of efficacious treatments for other forms of HyOb. This review aims to summarize our current understanding of the neuroendocrine physiology of appetite and weight regulation, and explore our current understanding of the pathophysiology of HyOb.
Collapse
Affiliation(s)
- Hoong-Wei Gan
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Manuela Cerbone
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Mehul Tulsidas Dattani
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
6
|
Choi PP, Wang Q, Brenner LA, Li AJ, Ritter RC, Appleyard SM. Lesion of NPY Receptor-expressing Neurons in Perifornical Lateral Hypothalamus Attenuates Glucoprivic Feeding. Endocrinology 2024; 165:bqae021. [PMID: 38368624 PMCID: PMC11043786 DOI: 10.1210/endocr/bqae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 01/19/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
Glucoprivic feeding is one of several counterregulatory responses (CRRs) that facilitates restoration of euglycemia following acute glucose deficit (glucoprivation). Our previous work established that glucoprivic feeding requires ventrolateral medullary (VLM) catecholamine (CA) neurons that coexpress neuropeptide Y (NPY). However, the connections by which VLM CA/NPY neurons trigger increased feeding are uncertain. We have previously shown that glucoprivation, induced by an anti-glycolygic agent 2-deoxy-D-glucose (2DG), activates perifornical lateral hypothalamus (PeFLH) neurons and that expression of NPY in the VLM CA/NPY neurons is required for glucoprivic feeding. We therefore hypothesized that glucoprivic feeding and possibly other CRRs require NPY-sensitive PeFLH neurons. To test this, we used the ribosomal toxin conjugate NPY-saporin (NPY-SAP) to selectively lesion NPY receptor-expressing neurons in the PeFLH of male rats. We found that NPY-SAP destroyed a significant number of PeFLH neurons, including those expressing orexin, but not those expressing melanin-concentrating hormone. The PeFLH NPY-SAP lesions attenuated 2DG-induced feeding but did not affect 2DG-induced increase in locomotor activity, sympathoadrenal hyperglycemia, or corticosterone release. The 2DG-induced feeding response was also significantly attenuated in NPY-SAP-treated female rats. Interestingly, PeFLH NPY-SAP lesioned male rats had reduced body weights and decreased dark cycle feeding, but this effect was not seen in female rats. We conclude that a NPY projection to the PeFLH is necessary for glucoprivic feeding, but not locomotor activity, hyperglycemia, or corticosterone release, in both male and female rats.
Collapse
Affiliation(s)
- Pique P Choi
- Neuroscience Program, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Qing Wang
- Neuroscience Program, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Lynne A Brenner
- Neuroscience Program, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Ai-Jun Li
- Neuroscience Program, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Robert C Ritter
- Neuroscience Program, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Suzanne M Appleyard
- Neuroscience Program, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
7
|
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
|
8
|
Kiive E, Kanarik M, Veidebaum T, Harro J. Neuropeptide Y gene variants and Agreeableness: interaction effect with the birth cohort and the serotonin transporter promoter polymorphism. Acta Neuropsychiatr 2024; 36:1-8. [PMID: 37070394 DOI: 10.1017/neu.2023.23] [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] [Indexed: 04/19/2023]
Abstract
OBJECTIVE Neuropeptide Y (NPY) is a powerful regulator of anxious states, including social anxiety, but evidence from human genetic studies is limited. Associations of common gene variants with behaviour have been described as subject to birth cohort effects, especially if the behaviour is socially motivated. This study aimed to examine the association of NPY rs16147 and rs5574 with personality traits in highly representative samples of two birth cohorts of young adults, the samples having been formed during a period of rapid societal transition. METHODS Both birth cohorts (original n = 1238) of the Estonian Children Personality Behaviour and Health Study (ECPBHS) self-reported personality traits of the five-factor model at 25 years of age. RESULTS A significant interaction effect of the NPY rs16147 and rs5574 and birth cohort on Agreeableness was found. The T/T genotype of NPY rs16147 resulted in low Agreeableness in the older cohort (born 1983) and in high Agreeableness in the younger cohort (born 1989). The C/C genotype of NPY rs5574 was associated with higher Agreeableness in the younger but not in the older cohort. In the NPY rs16147 T/T homozygotes, the deviations from average in Agreeableness within the birth cohort were dependent on the serotonin transporter promoter polymorphism. CONCLUSIONS The association between the NPY gene variants and a personality domain reflecting social desirability is subject to change qualitatively in times of rapid societal changes, serving as an example of the relationship between the plasticity genes and environment. The underlying mechanism may involve the development of the serotonergic system.
Collapse
Affiliation(s)
- Evelyn Kiive
- Division of Special Education, Department of Education, University of Tartu, Jakobi 5, 51005 Tartu, Estonia
| | - Margus Kanarik
- Division of Neuropsychopharmacology, Department of Chemistry, University of Tartu, Ravila 14A, 50411 Tartu, Estonia
| | - Toomas Veidebaum
- Department of Chronic Diseases, National Institute for Health Development, Hiiu 42, 11619 Tallinn, Estonia
| | - Jaanus Harro
- Division of Neuropsychopharmacology, Department of Chemistry, University of Tartu, Ravila 14A, 50411 Tartu, Estonia
| |
Collapse
|
9
|
Mahdavi K, Zendehdel M, Baghbanzadeh A. The effects of neuropeptide W on food consumption and feeding behavior in neonatal meat-type chicks: Role of CRF1/CRF2 and NPY1 receptors. Neurosci Lett 2023; 817:137531. [PMID: 37863422 DOI: 10.1016/j.neulet.2023.137531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 10/17/2023] [Indexed: 10/22/2023]
Abstract
In several studies, the regulatory role of the neuropeptide W (NPW) system in food intake has been demonstrated. Considering the lack of avian studies in this field, the current research was conducted to evaluate the effects of intracerebroventricular (ICV) infusion of NPW and its interferences with corticotropin, melanocortin, and neuropeptide Y (NPY) receptors on meal consumption and feeding behaviors of broilers. In the first experiment, birds were injected with NPW (0.75, 1.5, and 3 nmol) in addition to saline. In the second experiment, saline, CRF1 receptor antagonist (NBI35965, 30 μg), NPW (3 nmol), and simultaneous injections of NBI35965 and NPW were performed. Experiments 3-8 were identical to experiment 2, except that CRF2 receptor antagonist (K41498, 30 μg), MC3/MC4 receptor antagonist (SHU9119, 0.5 nmol), MC4 receptor antagonist (HS024, 0.5 nmol), NPY1 receptor antagonist (BMS193885, 1.25 nmol), NPY2 receptor antagonist (CYM9484, 1.25 nmol), and NPY5 receptor (antagonist L-152,804, 1.25 nmol) were administrated instead of NBI35965. After that, cumulative feed intake and feeding behavior were monitored for 2 h and 30 min after injections, respectively. Following the infusion of NPW (1.5 and 3 nmol), there was a significant stimulation of meal consumption in chickens (P < 0.05). Concomitant injection of NBI35965 and K41498 with NPW enhanced the appetite-increasing effect of NPW (P < 0.05); while BMS193885 suppressed this effect of NPW (P < 0.05). Injection of SHU9119, HS024, CYM9484, and L-152804 with NPW at the same time, had no significant effect on NPW-induced hyperphagia (P > 0.05). NPW also significantly decreased the standing period and the number of jumps, steps, and exploratory pecks, and led to an increase in sitting period and feeding pecks (P < 0.05). Based on the observations, it seems that NPW-induced hyperphagia could be mediated through CRF1, CRF2, and NPY1 receptors in neonatal broilers.
Collapse
Affiliation(s)
- Kimia Mahdavi
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, 14155-6453 Tehran, Iran
| | - Morteza Zendehdel
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, 14155-6453 Tehran, Iran.
| | - Ali Baghbanzadeh
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, 14155-6453 Tehran, Iran
| |
Collapse
|
10
|
Lee YH, Kim YB, Kim KS, Jang M, Song HY, Jung SH, Ha DS, Park JS, Lee J, Kim KM, Cheon DH, Baek I, Shin MG, Lee EJ, Kim SJ, Choi HJ. Lateral hypothalamic leptin receptor neurons drive hunger-gated food-seeking and consummatory behaviours in male mice. Nat Commun 2023; 14:1486. [PMID: 36932069 PMCID: PMC10023672 DOI: 10.1038/s41467-023-37044-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/01/2023] [Indexed: 03/19/2023] Open
Abstract
For survival, it is crucial for eating behaviours to be sequenced through two distinct seeking and consummatory phases. Heterogeneous lateral hypothalamus (LH) neurons are known to regulate motivated behaviours, yet which subpopulation drives food seeking and consummatory behaviours have not been fully addressed. Here, in male mice, fibre photometry recordings demonstrated that LH leptin receptor (LepR) neurons are correlated explicitly in both voluntary seeking and consummatory behaviours. Further, micro-endoscope recording of the LHLepR neurons demonstrated that one subpopulation is time-locked to seeking behaviours and the other subpopulation time-locked to consummatory behaviours. Seeking or consummatory phase specific paradigm revealed that activation of LHLepR neurons promotes seeking or consummatory behaviours and inhibition of LHLepR neurons reduces consummatory behaviours. The activity of LHLepR neurons was increased via Neuropeptide Y (NPY) which acted as a tonic permissive gate signal. Our results identify neural populations that mediate seeking and consummatory behaviours and may lead to therapeutic targets for maladaptive food seeking and consummatory behaviours.
Collapse
Affiliation(s)
- Young Hee Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Yu-Been Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Kyu Sik Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Mirae Jang
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Physiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Ha Young Song
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Sang-Ho Jung
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Dong-Soo Ha
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Joon Seok Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Jaegeon Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Physiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Kyung Min Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Deok-Hyeon Cheon
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Inhyeok Baek
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Min-Gi Shin
- Department of Brain Science, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
| | - Eun Jeong Lee
- Department of Brain Science, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
| | - Sang Jeong Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Physiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Wide River Institute of Immunology, Seoul National University, 101 Dabyeonbat-gil, Hwachon-myeon, Gangwon-do, 25159, Republic of Korea
| | - Hyung Jin Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Neuroscience Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Wide River Institute of Immunology, Seoul National University, 101 Dabyeonbat-gil, Hwachon-myeon, Gangwon-do, 25159, Republic of Korea.
| |
Collapse
|
11
|
Singh U, Alex R, Chaudhary A, Deb R, Raja TV, Rathod B, Savaliya BD, Kumar S, Das AK. Genetic variants in 5'UTR and exonic region of NPY gene alter the reproduction performance in Indian cattle breeds. Reprod Domest Anim 2023; 58:246-252. [PMID: 36269691 DOI: 10.1111/rda.14281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/11/2022] [Accepted: 10/15/2022] [Indexed: 11/29/2022]
Abstract
Neuropeptide Y (NPY) is one of the most potent orexigenic factors which can produce diverse effects on behaviour and other physiological functions and is highly conserved in evolution. The present study was aimed to identify and associate SNPs in the 5' UTR and exon2 region of the NPY gene with reproduction and production traits in Kankrej cattle of Indian origin. Three mutations in the 5'-UTR region and one mutation in the exon2 region of the NPY gene were identified by PCR-SSCP and PCR-RFLP, respectively, followed by sequencing. Further, association studies were conducted with reproduction and production traits in Kankrej cattle. The GACCGA genotyped animals based on the 5'UTR variants indicated better dry period and calving interval, whereas with GGCCGG genotypes showed higher total lactation milk yield and 305-day milk yield in comparison to other genotypes. Also, service period and inter calving period varied significantly among the genotypes of exon2, as the GG genotyped animals had significantly longer calving interval. Other traits like age at first heat, age at first service and age at first calving were not affected by the mutations. So, the present study outlined that the bovine NPY gene may be considered to be one of the candidate gene for improvement of reproductive performance of cattle, after validation on large sample size.
Collapse
Affiliation(s)
- Umesh Singh
- ICAR-Central Institute for Research on Cattle, Meerut, India
| | - Rani Alex
- ICAR-Central Institute for Research on Cattle, Meerut, India
| | | | - Rajib Deb
- ICAR-Central Institute for Research on Cattle, Meerut, India
| | | | - Bharatsingh Rathod
- Livestock Research Station, Sardarkrushinagar Dantiwada Agricultural University, Banaskatha, India
| | | | - Sushil Kumar
- ICAR-Central Institute for Research on Cattle, Meerut, India
| | | |
Collapse
|
12
|
Liu Y, Gu R, Gao M, Wei Y, Shi Y, Wang X, Gu Y, Gu X, Zhang H. Emerging role of substance and energy metabolism associated with neuroendocrine regulation in tumor cells. Front Endocrinol (Lausanne) 2023; 14:1126271. [PMID: 37051193 PMCID: PMC10084767 DOI: 10.3389/fendo.2023.1126271] [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: 12/17/2022] [Accepted: 02/07/2023] [Indexed: 03/29/2023] Open
Abstract
Cancer is the second most common cause of mortality in the world. One of the unresolved difficult pathological mechanism issues in malignant tumors is the imbalance of substance and energy metabolism of tumor cells. Cells maintain life through energy metabolism, and normal cells provide energy through mitochondrial oxidative phosphorylation to generate ATP, while tumor cells demonstrate different energy metabolism. Neuroendocrine control is crucial for tumor cells' consumption of nutrients and energy. As a result, better combinatorial therapeutic approaches will be made possible by knowing the neuroendocrine regulating mechanism of how the neuroendocrine system can fuel cellular metabolism. Here, the basics of metabolic remodeling in tumor cells for nutrients and metabolites are presented, showing how the neuroendocrine system regulates substance and energy metabolic pathways to satisfy tumor cell proliferation and survival requirements. In this context, targeting neuroendocrine regulatory pathways in tumor cell metabolism can beneficially enhance or temper tumor cell metabolism and serve as promising alternatives to available treatments.
Collapse
Affiliation(s)
- Yingying Liu
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
- School of Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Renjun Gu
- School of Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Murong Gao
- Beijing Rehabilitation Hospital Affiliated to Capital Medical University, Beijing, China
| | - Yangwa Wei
- Department of Hepatobiliary Surgery, Hainan Provincial People’s Hospital, Haikou, China
| | - Yu Shi
- Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xu Wang
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yihuang Gu
- School of Acupuncture and Tuina, School of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
- The Second Hospital of Nanjing, Nanjing, China
- *Correspondence: Hongru Zhang, ; Xin Gu, ; Yihuang Gu,
| | - Xin Gu
- School of Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Hongru Zhang, ; Xin Gu, ; Yihuang Gu,
| | - Hongru Zhang
- School of Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Hongru Zhang, ; Xin Gu, ; Yihuang Gu,
| |
Collapse
|
13
|
Zhao X, Si L, Niu L, Wei M, Wang F, Liu X, Chen Z, Qiao Y, Cheng L, Yang S. Effects of RFRP‑3 on an ovariectomized estrogen‑primed rat model and HEC‑1A human endometrial carcinoma cells. Exp Ther Med 2022; 25:76. [PMID: 36684658 PMCID: PMC9842939 DOI: 10.3892/etm.2022.11775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 11/10/2022] [Indexed: 12/24/2022] Open
Abstract
The hypothalamic peptide gonadotropin inhibitory hormone (GnIH) is a relatively novel hypothalamic neuropeptide, identified in 2000. It can influence the hypothalamic-pituitary-gonadal axis and reproductive function through various neuroendocrine systems. The present study aimed to explore the effects and potential underlying molecular mechanism of RFamide-related peptide-3 (RFRP-3) injection on the uterine fluid protein profile of ovariectomized estrogen-primed (OEP) rats using proteomics. In addition, the possible effects of RFRP-3 on the viability and apoptosis of the human endometrial cancer cell line HEC-1A and associated molecular mechanism were investigated. The OEP rat model was established through injection with GnIH/RFRP-3 through the lateral ventricle. At 6 h after injection, the protein components of uterine fluid of rats in the experimental and control groups were analyzed using liquid chromatography (LC)-tandem mass spectrometry (MS/MS). Differentially expressed proteins (DEPs) were analyzed using the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases. Protein-protein interactions (PPI) were investigated using the STRING database. PPI networks were then established before hub proteins were selected using OmicsBean software. The expression of one of the hub proteins, Kras, was then detected using western blot analysis. Cell Counting Kit-8, Annexin V-FITC/PI, reverse transcription-quantitative PCR and western blotting were also performed to analyze cell viability and apoptosis. In total, 417 DEPs were obtained using LC-MS/MS, including 279 upregulated and 138 downregulated proteins. GO analysis revealed that the majority of the DEPs were secretory proteins. According to KEGG enrichment analysis, the DEPs found were generally involved in tumor-associated pathways. In particular, five hub proteins, namely G protein subunit α (Gna)13, Gnaq, Gnai3, Kras and MMP9, were obtained following PPI network analysis. Western blot analysis showed that expression of the hub protein Kras was downregulated following treatment with 10,000 ng/ml RFRP-3. RFRP-3 treatment (10,000 ng/ml) also suppressed HEC-1A cell viability, induced apoptosis, downregulated Bcl-2 and upregulated Bax protein expression, compared with those in the control group. In addition, compared with those in the control group, RFRP-3 significantly reduced the mRNA expression levels of PI3K, AKT and mTOR, while upregulating those of LC3-II. Compared with those in the control group, RFRP-3 significantly decreased the protein expression levels of PI3K, AKT, mTOR and p62, in addition to decreasing AKT phosphorylation. By contrast, RFRP-3 significantly increased the LC3-II/I ratio and G protein-coupled receptor 147 (GPR147) protein expression. In conclusion, the present data suggest that RFRP-3 can alter the protein expression profile of the uterine fluid of OEP rats by upregulating MMP9 expression whilst downregulating that of key hub proteins Gna13, GnaQ, Gnai3 and Kras. Furthermore, RFRP-3 can inhibit HEC-1A cell viability while promoting apoptosis. The underlying molecular mechanism may involve activation of GPR147 receptor by the direct binding of RFRP-3, which further downregulates the hub protein Kras to switch on the PI3K/AKT/mTOR pathway. This subsequently reduces the Bcl-2 expression and promotes Bax expression to induce autophagy.
Collapse
Affiliation(s)
- Xueying Zhao
- Department of Immunology, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Lina Si
- Department of Human Anatomy, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Lin Niu
- Department of Human Anatomy, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Meng Wei
- Department of Human Anatomy, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Fengxia Wang
- Department of Human Anatomy, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Xiaochao Liu
- Department of Human Anatomy, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Zhihong Chen
- Department of Human Anatomy, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Yuebing Qiao
- Department of Human Anatomy, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Luyang Cheng
- Department of Immunology, Chengde Medical University, Chengde, Hebei 067000, P.R. China,Correspondence to: Mrs. Luyang Cheng, Department of Immunology, Chengde Medical University, Anyuan Road, Shuangqiao, Chengde, Hebei 067000, P.R. China
| | - Songhe Yang
- Department of Human Anatomy, Chengde Medical University, Chengde, Hebei 067000, P.R. China,Correspondence to: Mrs. Luyang Cheng, Department of Immunology, Chengde Medical University, Anyuan Road, Shuangqiao, Chengde, Hebei 067000, P.R. China
| |
Collapse
|
14
|
Ceccarini MR, Precone V, Manara E, Paolacci S, Maltese PE, Benfatti V, Dhuli K, Donato K, Guerri G, Marceddu G, Chiurazzi P, Dalla Ragione L, Beccari T, Bertelli M. A next generation sequencing gene panel for use in the diagnosis of anorexia nervosa. Eat Weight Disord 2022; 27:1869-1880. [PMID: 34822136 DOI: 10.1007/s40519-021-01331-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/07/2021] [Indexed: 12/12/2022] Open
Abstract
PURPOSE The aim of this study was to increase knowledge of genes associated with anorexia nervosa (AN) and their diagnostic offer, using a next generation sequencing (NGS) panel for the identification of genetic variants. The rationale underlying this test is that we first analyze the genes associated with syndromic forms of AN, then genes that were found to carry rare variants in AN patients who had undergone segregation analysis, and finally candidate genes intervening in the same molecular pathways or identified by GWAS or in mouse models. METHODS We developed an NGS gene panel and used it to screen 68 Italian AN patients (63 females, 5 males). The panel included 162 genes. Family segregation study was conducted on available relatives of probands who reported significant genetic variants. RESULTS In our analysis, we found potentially deleterious variants in 2 genes (PDE11A and SLC25A13) associated with syndromic forms of anorexia and predicted deleterious variants in the following 12 genes: CD36, CACNA1C, DRD4, EPHX2, ESR1, GRIN2A, GRIN3B, LRP2, NPY4R, PTGS2, PTPN22 and SGPP2. Furthermore, by Sanger sequencing of the promoter region of NNAT, we confirmed the involvement of this gene in the pathogenesis of AN. Family segregation studies further strengthened the possible causative role of CACNA1C, DRD4, GRIN2A, PTGS2, SGPP2, SLC25A13 and NNAT genes in AN etiology. CONCLUSION The major finding of our study is the confirmation of the involvement of the NNAT gene in the pathogenesis of AN; furthermore, this study suggests that NGS-based testing can play an important role in the diagnostic evaluation of AN, excluding syndromic forms and increasing knowledge of the genetic etiology of AN. LEVEL OF EVIDENCE Level I, experimental study.
Collapse
Affiliation(s)
- Maria Rachele Ceccarini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy.
- C.I.B., Consorzio Interuniversitario per le Biotecnologie, Trieste, Italy.
| | | | | | | | | | - Valentina Benfatti
- Department of Eating Disorder, Palazzo Francisci Todi, USL 1 Umbria, Todi, PG, Italy
| | | | | | | | | | - Pietro Chiurazzi
- Dipartimento Universitario Scienze della Vita e Sanità Pubblica, Sezione di Medicina Genomica, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, UOC Genetica Medica, 00168, Roma, Italy
| | - Laura Dalla Ragione
- Department of Eating Disorder, Palazzo Francisci Todi, USL 1 Umbria, Todi, PG, Italy
- Food Science and Human Nutrition Unit, University Campus Biomedico of Rome, Rome, Italy
| | - Tommaso Beccari
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
- C.I.B., Consorzio Interuniversitario per le Biotecnologie, Trieste, Italy
| | | |
Collapse
|
15
|
Waddell IS, Orfila C. Dietary fiber in the prevention of obesity and obesity-related chronic diseases: From epidemiological evidence to potential molecular mechanisms. Crit Rev Food Sci Nutr 2022; 63:8752-8767. [PMID: 35471164 DOI: 10.1080/10408398.2022.2061909] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Obesity is a mostly preventable diet-related disease and currently a major challenge for human populations worldwide. Obesity is a major risk factor for diseases such as type 2 diabetes mellitus (T2DM), cardiovascular disease (CVD) and certain cancers. Dietary fiber is a complex mixture of non-digestible molecules, mostly polysaccharides. Multiple epidemiological studies have demonstrated statistically significant reductions in risks of obesity, T2DM, CVD, colorectal cancer, and pre-menopausal breast cancer with higher dietary fiber intakes. Various direct and indirect mechanisms have been proposed including altered digestion and absorption, stimulation of gut hormones including glucagon-like-peptide-1 (GLP-1) and peptide YY (PYY), reduced appetite, and altered metabolism of bile and cholesterol. These may act via pathways involving G-protein-coupled receptors (GPRs), histone deacetylase (HDAC), and aromatase enzymes. Ultimately, fiber intake contributes to improving glucose levels and insulin sensitivity, lowering risk of T2DM, CVD and certain cancers. Therefore, diets rich in dietary fiber should be encouraged to prevent obesity and associated chronic disease.
Collapse
Affiliation(s)
- Isabella Skye Waddell
- School of Food Science and Nutrition, Woodhouse Lane, University of Leeds, Leeds, UK
| | - Caroline Orfila
- School of Food Science and Nutrition, Woodhouse Lane, University of Leeds, Leeds, UK
| |
Collapse
|
16
|
Involvement of Neuropeptide Y within Paraventricular Nucleus in Electroacupuncture Inhibiting Sympathetic Activities in Hypertensive Rats. Int J Hypertens 2022; 2022:9990854. [PMID: 35087687 PMCID: PMC8789434 DOI: 10.1155/2022/9990854] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/05/2022] [Indexed: 12/14/2022] Open
Abstract
Although electroacupuncture (EA) has been used to decrease the blood pressure (BP) clinically, the underlying mechanisms are not clearly clarified. This study aimed to assess the hypothesis that EA treatment exerts a hypotensive action via suppressing sympathetic activities and modulating neuropeptide Y (NPY) function within the paraventricular nucleus (PVN) of hypertensive rats. Male Sprague-Dawley rats were selected for the experiment, and the hypertensive models were established by the two-kidney, one-clip (2K1C) method. Then, the rats were randomly assigned to the sham group, 2K1C group, 2K1C plus EA group, and 2K1C plus sham EA group. EA treatment at the acupoints ST36 and ST40 overlying the peroneal nerves was given once a day for 30 days. The radiotelemetry system was applied to collect the arterial BP recordings. Power spectral analyses of BP variability, BP responses to ganglionic blockade, and plasma levels of norepinephrine and epinephrine were performed to assess the changes in sympathetic nerve activity. Real-time PCR and Western blots were carried out to examine the expression of NPY system in the PVN. The responses of PVN microinjection with NPY Y1R antagonist BIBO3304 were detected to check the endogenous NPY tone. The results showed that the enhanced arterial BP and sympathetic activities were effectively reduced by 30 days of EA treatment, and baroreflex sensitivity was improved in 2K1C hypertensive rats. The level of NPY mRNA and protein expression in the PVN was markedly upregulated by EA treatment in 2K1C rats. In addition, the pressor responses of PVN microinjection with NPY Y1R antagonist BIBO3304 in 2K1C models were remarkably augmented by the EA stimulation. Our results indicate that the increased NPY expression and function in the PVN induced by EA treatment contribute to antihypertensive and sympathetic suppression on hypertensive rats. The findings may elucidate the underlying mechanisms of the acupuncture to be a potential therapeutic strategy against hypertension.
Collapse
|
17
|
Yamada S, van Kooten N, Mori T, Taguchi K, Tsujimura A, Tanaka M. Efferent and Afferent Connections of Neuropeptide Y Neurons in the Nucleus Accumbens of Mice. Front Neuroanat 2021; 15:741868. [PMID: 34566585 PMCID: PMC8460764 DOI: 10.3389/fnana.2021.741868] [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: 07/15/2021] [Accepted: 08/16/2021] [Indexed: 11/23/2022] Open
Abstract
Neuropeptide Y (NPY) is a neural peptide distributed widely in the brain and has various functions in each region. We previously reported that NPY neurons in the nucleus accumbens (NAc) are involved in the regulation of anxiety behavior. Anterograde and retrograde tracing studies suggest that neurons in the NAc project to several areas, such as the lateral hypothalamus (LH) and ventral pallidum (VP), and receive afferent projections from the cortex, thalamus, and amygdala. However, the neural connections between accumbal NPY neurons and other brain areas in mice remain unclear. In this study, we sought to clarify these anatomical connections of NPY neurons in the NAc by investigating their neural outputs and inputs. To selectively map NPY neuronal efferents from the NAc, we injected Cre-dependent adeno-associated viruses (AAVs) into the NAc of NPY-Cre mice. This revealed that NAc NPY neurons exclusively projected to the LH. We confirmed this by injecting cholera toxin b subunit (CTb), a retrograde tracer, into the LH and found that approximately 7–10% of NPY neurons in the NAc were double-labeled for mCherry and CTb. Moreover, retrograde tracing using recombinant rabies virus (rRABV) also identified NAc NPY projections to the LH. Finally, we investigated monosynaptic input to the NPY neurons in the NAc using rRABV. We found that NPY neurons in the NAc received direct synaptic connections from the midline thalamic nuclei and posterior basomedial amygdala. These findings provide new insight into the neural networks of accumbal NPY neurons and should assist in elucidating their functional roles.
Collapse
Affiliation(s)
- Shunji Yamada
- Department of Anatomy and Neurobiology, Graduate School of Medical, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Nienke van Kooten
- Department of Anatomy and Neurobiology, Graduate School of Medical, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takuma Mori
- Department of Molecular and Cellular Physiology Shinshu University, School of Medicine, Matsumoto, Japan
| | - Katsutoshi Taguchi
- Department of Anatomy and Neurobiology, Graduate School of Medical, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Atsushi Tsujimura
- Department of Basic Geriatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masaki Tanaka
- Department of Anatomy and Neurobiology, Graduate School of Medical, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
18
|
Kamyshna II, Pavlovych LB, Maslyanko VA, Kamyshnyi AM. Analysis of the transcriptional activity of genes of neuropeptides and their receptors in the blood of patients with thyroid pathology. J Med Life 2021; 14:243-249. [PMID: 34104248 PMCID: PMC8169137 DOI: 10.25122/jml-2020-0183] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The thyroid hormone plays a vital role in the development and maturation of the nervous system not only during prenatal and perinatal age but also in adults. “Peripheral marker hypothesis” revealed that gene expression changes in some regions of the brain are reflected into the peripheral blood lymphocytes. The objective of the study was to investigate changes in the gene expression profile of neuropeptides and their receptors in patients with different forms of thyroid pathology. One hundred fifty-three patients with thyroid pathology were enrolled in the study. They were divided into three groups: group 1 included 16 patients with postoperative hypothyroidism, group 2 included 65 patients with hypothyroidism resulting from autoimmune thyroiditis (AIT), and group 3 included 72 patients with AIT and elevated levels of anti-thyroglobulin (anti-Tg) and anti-thyroid peroxidase (anti-TPO) antibodies in the serum. We used a pathway-specific polymerase chain reaction (PCR) array (RT2 Profiler™ PCR Array Human Neurotrophins & Receptors, QIAGEN, Germany) to identify and verify neuropeptides and receptors pathway-focused gene expression in 12 individuals that were randomly selected from each group using real-time PCR. Our research identified that patients with postoperative hypothyroidism had a considerably increased expression of NPY1R, NTSR1, and NPY4R. The patients with hypothyroidism caused by autoimmune thyroiditis had considerably lower expression of NTSR1, while the expression of NPY1R increased. The mRNA levels of NPY2R and PNOC increased in the patients with elevated levels of autoantibodies anti-Tg and anti-TPO in the serum, and mRNA levels of NPY1R and NTSR1 decreased in this group of patients.
Collapse
Affiliation(s)
- Iryna Ivanivna Kamyshna
- Department of Medical Rehabilitation, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Larysa Borysivna Pavlovych
- Department of Clinical Immunology, Allergology and Endocrinology, HSEEU Bukovinian State Medical University, Chernivtsi, Ukraine
| | - Vitaliy Antonovych Maslyanko
- Department of Clinical Immunology, Allergology and Endocrinology, HSEEU Bukovinian State Medical University, Chernivtsi, Ukraine
| | | |
Collapse
|
19
|
Endocrine role of bone in the regulation of energy metabolism. Bone Res 2021; 9:25. [PMID: 34016950 PMCID: PMC8137703 DOI: 10.1038/s41413-021-00142-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 12/20/2020] [Accepted: 01/12/2021] [Indexed: 02/06/2023] Open
Abstract
Bone mainly functions as a supportive framework for the whole body and is the major regulator of calcium homeostasis and hematopoietic function. Recently, an increasing number of studies have characterized the significance of bone as an endocrine organ, suggesting that bone-derived factors regulate local bone metabolism and metabolic functions. In addition, these factors can regulate global energy homeostasis by altering insulin sensitivity, feeding behavior, and adipocyte commitment. These findings may provide a new pathological mechanism for related metabolic diseases or be used in the diagnosis, treatment, and prevention of metabolic diseases such as osteoporosis, obesity, and diabetes mellitus. In this review, we summarize the regulatory effect of bone and bone-derived factors on energy metabolism and discuss directions for future research.
Collapse
|
20
|
Xu S, Yang H, Menon V, Lemire AL, Wang L, Henry FE, Turaga SC, Sternson SM. Behavioral state coding by molecularly defined paraventricular
hypothalamic cell type ensembles. Science 2020; 370:370/6514/eabb2494. [DOI: 10.1126/science.abb2494] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 08/18/2020] [Indexed: 12/11/2022]
Abstract
Brains encode behaviors using neurons amenable to systematic
classification by gene expression. The contribution of molecular identity to
neural coding is not understood because of the challenges involved with
measuring neural dynamics and molecular information from the same cells. We
developed CaRMA (calcium and RNA multiplexed activity) imaging based on
recording in vivo single-neuron calcium dynamics followed by gene expression
analysis. We simultaneously monitored activity in hundreds of neurons in
mouse paraventricular hypothalamus (PVH). Combinations of cell-type marker
genes had predictive power for neuronal responses across 11 behavioral
states. The PVH uses combinatorial assemblies of molecularly defined neuron
populations for grouped-ensemble coding of survival behaviors. The
neuropeptide receptor neuropeptide Y receptor type 1 (Npy1r) amalgamated
multiple cell types with similar responses. Our results show that
molecularly defined neurons are important processing units for brain
function.
Collapse
Affiliation(s)
- Shengjin Xu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Hui Yang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Vilas Menon
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Andrew L. Lemire
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Lihua Wang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Fredrick E. Henry
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Srinivas C. Turaga
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Scott M. Sternson
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| |
Collapse
|
21
|
Gumbs MCR, Eggels L, Kool T, Unmehopa UA, van den Heuvel JK, Lamuadni K, Mul JD, la Fleur SE. Neuropeptide Y Signaling in the Lateral Hypothalamus Modulates Diet Component Selection and is Dysregulated in a Model of Diet-Induced Obesity. Neuroscience 2019; 447:28-40. [PMID: 31887359 DOI: 10.1016/j.neuroscience.2019.12.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 12/06/2019] [Accepted: 12/09/2019] [Indexed: 01/16/2023]
Abstract
The preclinical multicomponent free-choice high-fat high-sucrose (fcHFHS) diet has strong validity to model diet-induced obesity (DIO) and associated maladaptive molecular changes in the central nervous system. fcHFHS-induced obese rats demonstrate increased sensitivity to intracerebroventricular infusion of the orexigenic Neuropeptide Y (NPY). The brain region-specific effects of NPY signaling on fcHFHS diet component selection are not completely understood. For example, fcHFHS-fed rats have increased intake of chow and fat following intracerebroventricular NPY infusion, whereas NPY administration in the nucleus accumbens, a key hub of the reward circuitry, specifically increases fat intake. Here, we investigated whether NPY infusion in the lateral hypothalamic area (LHA), which is crucially involved in the regulation of intake, regulates fcHFHS component selection, and if LHA NPY receptor subtypes 1 or 5 (NPYR1/5) are involved. Male Wistar rats were fed a chow or fcHFHS diet for at least seven days, and received intra-LHA vehicle or NPY infusions in a cross-over design. Diet component intake was measured two hours later. Separate experimental designs were used to test the efficacy of NPY1R- or NPY5R antagonism to prevent the orexigenic effects of intra-LHA NPY. Intra-LHA NPY increased caloric intake in chow- and fcHFHS-fed rats. This effect was mediated specifically by chow intake in fcHFHS-fed rats. The orexigenic effects of intra-LHA NPY were prevented by NPY1R and NPY5R antagonism in chow-fed rats, but only by NPY5R antagonism in fcHFHS-fed rats. Thus, NPY signaling has brain region-specific effects on fcHFHS component selection and LHA NPYR sensitivity is dysregulated during consumption of a fcHFHS diet.
Collapse
Affiliation(s)
- M C R Gumbs
- Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism and Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; Metabolism and Reward Group, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| | - L Eggels
- Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism and Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; Metabolism and Reward Group, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| | - T Kool
- Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism and Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - U A Unmehopa
- Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism and Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - J K van den Heuvel
- Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism and Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - K Lamuadni
- Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism and Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; Metabolism and Reward Group, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| | - J D Mul
- Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism and Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; Metabolism and Reward Group, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Sciencepark 904, 1098 XH Amsterdam, The Netherlands
| | - S E la Fleur
- Amsterdam UMC, University of Amsterdam, Department of Endocrinology and Metabolism and Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; Metabolism and Reward Group, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Meibergdreef 47, 1105 BA Amsterdam, The Netherlands.
| |
Collapse
|
22
|
Comeras LB, Herzog H, Tasan RO. Neuropeptides at the crossroad of fear and hunger: a special focus on neuropeptide Y. Ann N Y Acad Sci 2019; 1455:59-80. [PMID: 31271235 PMCID: PMC6899945 DOI: 10.1111/nyas.14179] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 05/15/2019] [Accepted: 06/03/2019] [Indexed: 12/11/2022]
Abstract
Survival in a natural environment forces an individual into constantly adapting purposive behavior. Specified interoceptive neurons monitor metabolic and physiological balance and activate dedicated brain circuits to satisfy essential needs, such as hunger, thirst, thermoregulation, fear, or anxiety. Neuropeptides are multifaceted, central components within such life‐sustaining programs. For instance, nutritional depletion results in a drop in glucose levels, release of hormones, and activation of hypothalamic and brainstem neurons. These neurons, in turn, release several neuropeptides that increase food‐seeking behavior and promote food intake. Similarly, internal and external threats activate neuronal pathways of avoidance and defensive behavior. Interestingly, specific nuclei of the hypothalamus and extended amygdala are activated by both hunger and fear. Here, we introduce the relevant neuropeptides and describe their function in feeding and emotional‐affective behaviors. We further highlight specific pathways and microcircuits, where neuropeptides may interact to identify prevailing homeostatic needs and direct respective compensatory behaviors. A specific focus will be on neuropeptide Y, since it is known for its pivotal role in metabolic and emotional pathways. We hypothesize that the orexigenic and anorexigenic properties of specific neuropeptides are related to their ability to inhibit fear and anxiety.
Collapse
Affiliation(s)
- Lucas B Comeras
- Department of Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| | - Herbert Herzog
- Neuroscience Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Ramon O Tasan
- Department of Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
23
|
Shebanits K, Andersson-Assarsson JC, Larsson I, Carlsson LMS, Feuk L, Larhammar D. Copy number of pancreatic polypeptide receptor gene NPY4R correlates with body mass index and waist circumference. PLoS One 2018; 13:e0194668. [PMID: 29621259 PMCID: PMC5886410 DOI: 10.1371/journal.pone.0194668] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/07/2018] [Indexed: 01/14/2023] Open
Abstract
Multiple genetic studies have linked copy number variation (CNV) in different genes to body mass index (BMI) and obesity. A CNV on chromosome 10q11.22 has been associated with body weight. This CNV region spans NPY4R, the gene encoding the pancreatic polypeptide receptor Y4, which has been described as a satiety-stimulating receptor. We have investigated CNV of the NPY4R gene and analysed its relationship to BMI, waist circumference and self-reported dietary intake from 558 individuals (216 men and 342 women) representing a wide BMI range. The copy number for NPY4R ranged from 2 to 8 copies (average 4.6±0.8). Rather than the expected negative correlation, we observed a positive correlation between NPY4R copy number and BMI as well as waist circumference in women (Pearson’s r = 0.267, p = 2.65×10−7 and r = 0.256, p = 8×10−7, respectively). Each additional copy of NPY4R correlated with 2.6 kg/m2 increase in BMI and 5.67 cm increase in waist circumference (p = 2.8×10−5 and p = 6.2×10−5, respectively) for women. For men, there was no statistically significant correlation between CNV and BMI. Our results suggest that NPY4R genetic variation influences body weight in women, but the exact role of this receptor appears to be more complex than previously proposed.
Collapse
Affiliation(s)
| | | | - Ingrid Larsson
- Dept. of Gastroenterology and Hepatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lena M. S. Carlsson
- Dept. of Molecular and Clinical Medicine, Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden
| | - Lars Feuk
- Dept. of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Dan Larhammar
- Dept. of Neuroscience, Uppsala University, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
24
|
Oki Y, Teraoka H, Kitazawa T. Neuropeptide Y (NPY) inhibits spontaneous contraction of the mouse atrium by possible activation of the NPY1 receptor. ACTA ACUST UNITED AC 2018; 37:23-28. [PMID: 28544313 DOI: 10.1111/aap.12055] [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: 01/14/2017] [Revised: 02/24/2017] [Accepted: 03/07/2017] [Indexed: 11/28/2022]
Abstract
Neuropeptide Y (NPY) causes various central and peripheral actions through activation of G-protein-coupled NPY receptors. Although a species-dependent difference in cardiac actions of NPY has been reported, the responses to NPY have not been examined in mice, widely used experimental animals. This study aimed to clarify the responses to NPY and the receptor subtype involved in the responses in mouse atrium. Neuropeptide Y caused negative inotropic and negative chronotropic actions in spontaneous beating right atria. Negative inotropic actions were more marked than negative chronotropic actions. Therefore, negative inotropic actions were studied in detail for evaluation of the NPY-induced cardiac actions in mouse atrium. Neuropeptide Y-induced negative inotropic actions were not affected by atropine but were abolished in the atria from pertussis toxin-treated mice. In isolated atrial preparations from reserpine-treated mice, NPY-induced negative inotropic actions were significantly attenuated. [Leu31, Pro34]-NPY, but not peptide YY, was effective in decreasing spontaneous contraction in atrial preparations. Although Y1 , Y2 , Y4 and Y5 receptor mRNAs were expressed almost equally in the brain, NPY1 receptor mRNA was dominantly expressed in the atrium. In conclusion, NPY caused negative inotropic and chronotropic actions through activation of the Y1 receptor in the mouse atrium. A high expression level of Y1 mRNA in the atrium suggests a functional role of NPY in the regulation of mouse cardiac contraction.
Collapse
Affiliation(s)
- Y Oki
- Department of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| | - H Teraoka
- Department of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| | - T Kitazawa
- Department of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| |
Collapse
|
25
|
Seldeen KL, Halley PG, Volmar CH, Rodríguez MA, Hernandez M, Pang M, Carlsson SK, Suva LJ, Wahlestedt C, Troen BR, Brothers SP. Neuropeptide Y Y2 antagonist treated ovariectomized mice exhibit greater bone mineral density. Neuropeptides 2018; 67:45-55. [PMID: 29129406 PMCID: PMC5805636 DOI: 10.1016/j.npep.2017.11.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 11/06/2017] [Accepted: 11/06/2017] [Indexed: 12/15/2022]
Abstract
Osteoporosis, a disease characterized by progressive bone loss and increased risk of fracture, often results from menopausal loss of estrogen in women. Neuropeptide Y has been shown to negatively regulate bone formation, with amygdala specific deletion of the Y2 receptor resulting in increased bone mass in mice. In this study, ovariectomized (OVX) mice were injected once daily with JNJ-31020028, a brain penetrant Y2 receptor small molecule antagonist to determine the effects on bone formation. Antagonist treated mice had reduced weight and showed increased whole-body bone mineral density compared to vehicle-injected mice. Micro computerized tomography (micro-CT) demonstrated increased vertebral trabecular bone volume, connectivity density and trabecular thickness. Femoral micro-CT analysis revealed increased bone volume within trabecular regions and greater trabecular number, without significant difference in other parameters or within cortical regions. A decrease was seen in serum P1NP, a measure used to confirm positive treatment outcomes in bisphosphonate treated patients. C-terminal telopeptide 1 (CTX-1), a blood biomarker of bone resorption, was decreased in treated animals. The higher bone mineral density observed following Y2 antagonist treatment, as determined by whole-body DEXA scanning, is indicative of either enhanced mineralization or reduced bone loss. Additionally, our findings that ex vivo treatment of bone marrow cells with the Y2 antagonist did not affect osteoblast and osteoclast formation suggests the inhibitor is not affecting these cells directly, and suggests a central role for compound action in this system. Our results support the involvement of Y2R signalling in bone metabolism and give credence to the hypothesis that selective pharmacological manipulation of Y2R may provide anabolic benefits for treating osteoporosis.
Collapse
Affiliation(s)
- K L Seldeen
- Division of Geriatrics and Palliative Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA; Research Service, Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA
| | - P G Halley
- Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - C H Volmar
- Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - M A Rodríguez
- Bruce W. Carter VA Geriatric Research Education and Clinical Center (GRECC), Miami, FL, USA; University of Miami Miller School of Medicine, Miami, FL, USA
| | - M Hernandez
- Bruce W. Carter VA Geriatric Research Education and Clinical Center (GRECC), Miami, FL, USA; University of Miami Miller School of Medicine, Miami, FL, USA
| | - M Pang
- Division of Geriatrics and Palliative Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA; Research Service, Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA
| | - S K Carlsson
- Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - L J Suva
- Department of Orthopaedic Surgery, Centre for Orthopaedic Research, University of Arkansas Medical School, Little Rock, AR, USA
| | - C Wahlestedt
- Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - B R Troen
- Division of Geriatrics and Palliative Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA; Research Service, Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA
| | - S P Brothers
- Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
26
|
Guarino D, Nannipieri M, Iervasi G, Taddei S, Bruno RM. The Role of the Autonomic Nervous System in the Pathophysiology of Obesity. Front Physiol 2017; 8:665. [PMID: 28966594 PMCID: PMC5606212 DOI: 10.3389/fphys.2017.00665] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 08/22/2017] [Indexed: 12/18/2022] Open
Abstract
Obesity is reaching epidemic proportions globally and represents a major cause of comorbidities, mostly related to cardiovascular disease. The autonomic nervous system (ANS) dysfunction has a two-way relationship with obesity. Indeed, alterations of the ANS might be involved in the pathogenesis of obesity, acting on different pathways. On the other hand, the excess weight induces ANS dysfunction, which may be involved in the haemodynamic and metabolic alterations that increase the cardiovascular risk of obese individuals, i.e., hypertension, insulin resistance and dyslipidemia. This article will review current evidence about the role of the ANS in short-term and long-term regulation of energy homeostasis. Furthermore, an increased sympathetic activity has been demonstrated in obese patients, particularly in the muscle vasculature and in the kidneys, possibily contributing to increased cardiovascular risk. Selective leptin resistance, obstructive sleep apnea syndrome, hyperinsulinemia and low ghrelin levels are possible mechanisms underlying sympathetic activation in obesity. Weight loss is able to reverse metabolic and autonomic alterations associated with obesity. Given the crucial role of autonomic dysfunction in the pathophysiology of obesity and its cardiovascular complications, vagal nerve modulation and sympathetic inhibition may serve as therapeutic targets in this condition.
Collapse
Affiliation(s)
- Daniela Guarino
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy.,Institute of Clinical Physiology of CNRPisa, Italy.,Scuola Superiore Sant'AnnaPisa, Italy
| | - Monica Nannipieri
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| | | | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| | - Rosa Maria Bruno
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| |
Collapse
|
27
|
Schubert M, Stichel J, Du Y, Tough IR, Sliwoski G, Meiler J, Cox HM, Weaver CD, Beck-Sickinger AG. Identification and Characterization of the First Selective Y4 Receptor Positive Allosteric Modulator. J Med Chem 2017; 60:7605-7612. [DOI: 10.1021/acs.jmedchem.7b00976] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Mario Schubert
- Faculty
of Biosciences, Pharmacy and Psychology, Institute of Biochemistry, Leipzig University, Leipzig 04103, Germany
| | - Jan Stichel
- Faculty
of Biosciences, Pharmacy and Psychology, Institute of Biochemistry, Leipzig University, Leipzig 04103, Germany
| | - Yu Du
- Department
of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Iain R. Tough
- Wolfson
Centre for Age-Related Diseases, King’s College London, Guy’s Campus, London SE1 1UL, U.K
| | - Gregory Sliwoski
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Jens Meiler
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
- Vanderbilt
Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Helen M. Cox
- Wolfson
Centre for Age-Related Diseases, King’s College London, Guy’s Campus, London SE1 1UL, U.K
| | - C. David Weaver
- Department
of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt
Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Annette G. Beck-Sickinger
- Faculty
of Biosciences, Pharmacy and Psychology, Institute of Biochemistry, Leipzig University, Leipzig 04103, Germany
| |
Collapse
|
28
|
Shi Z, Madden CJ, Brooks VL. Arcuate neuropeptide Y inhibits sympathetic nerve activity via multiple neuropathways. J Clin Invest 2017. [PMID: 28628036 PMCID: PMC5490747 DOI: 10.1172/jci92008] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Obesity increases sympathetic nerve activity (SNA) via activation of proopiomelanocortin neurons in the arcuate nucleus (ArcN), and this action requires simultaneous withdrawal of tonic neuropeptide Y (NPY) sympathoinhibition. However, the sites and neurocircuitry by which NPY decreases SNA are unclear. Here, using designer receptors exclusively activated by designer drugs (DREADDs) to selectively activate or inhibit ArcN NPY neurons expressing agouti-related peptide (AgRP) in mice, we have demonstrated that this neuronal population tonically suppresses splanchnic SNA (SSNA), arterial pressure, and heart rate via projections to the paraventricular nucleus (PVN) and dorsomedial hypothalamus (DMH). First, we found that ArcN NPY/AgRP fibers closely appose PVN and DMH presympathetic neurons. Second, nanoinjections of NPY or an NPY receptor Y1 (NPY1R) antagonist into PVN or DMH decreased or increased SSNA, respectively. Third, blockade of DMH NPY1R reversed the sympathoinhibition elicited by selective, DREADD-mediated activation of ArcN NPY/AgRP neurons. Finally, stimulation of ArcN NPY/AgRP terminal fields in the PVN and DMH decreased SSNA. Considering that chronic obesity decreases ArcN NPY content, we propose that the ArcN NPY neuropathway to the PVN and DMH is pivotal in obesity-induced elevations in SNA.
Collapse
Affiliation(s)
- Zhigang Shi
- Department of Physiology and Pharmacology and
| | - Christopher J Madden
- Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon, USA
| | | |
Collapse
|
29
|
Enkephalin and neuropeptide-Y interaction in the intergeniculate leaflet network, a part of the mammalian biological clock. Neuroscience 2017; 343:10-20. [DOI: 10.1016/j.neuroscience.2016.11.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/22/2016] [Accepted: 11/22/2016] [Indexed: 11/20/2022]
|
30
|
Murase SI, Shiiya T, Higuchi H. Neuropeptide Y Y 5 receptor localization in mouse central nervous system. Brain Res 2017; 1655:216-232. [PMID: 27984021 DOI: 10.1016/j.brainres.2016.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 10/12/2016] [Accepted: 10/27/2016] [Indexed: 10/20/2022]
Abstract
Neuropeptide Y (NPY) and its receptors affect blood pressure, feeding behavior, and neurogenesis. In this study, the distribution of neurons expressing NPY Y5 receptor (Y5) was examined in adult mouse central nervous system by immunohistochemistry. Y5 protein localization was investigated using polyclonal anti-Y5 antibody, which was successfully preabsorbed with Y5 knockout brain tissues. The preabsorbed anti-Y5 antibody did not react with Y5 knockout brain tissues, thus meeting the "hard specificity criterion," which is the absence of staining in tissues genetically deficient for the antigen (Pradidarcheep et al., 2008). Y5-positive neurons were found in most brain areas. Most Y5 immunoreactivities were observed as dot-like structures adjacent to the plasma membrane, as expected for a cell membrane receptor. In situ hybridization showed that the Y5 mRNA expression was correlated with the Y5 protein level in each case and that it was probably controlled by the transcriptional regulation of the Y5 gene. In the nuclei where Y5 was expressed, Y5 immunoreactivities were found mainly in the somatic and dendritic areas. The distribution patterns of the Y5-positive cells that were broader than previously expected suggest important biological activities of the Y5 in many brain areas.
Collapse
Affiliation(s)
- Shin-Ichi Murase
- Division of Pharmacology, Niigata University, Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan.
| | - Tomohiro Shiiya
- Division of Pharmacology, Niigata University, Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Hiroshi Higuchi
- Division of Pharmacology, Niigata University, Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan.
| |
Collapse
|
31
|
Domin H, Święch D, Piergies N, Pięta E, Kim Y, Proniewicz E. Characterization of the surface geometry of acetyl-[Leu 28,31 ]-NPY(24-36), a selective Y 2 receptor agonist, onto the Ag and Au surfaces. VIBRATIONAL SPECTROSCOPY 2016; 85:1-6. [DOI: 10.1016/j.vibspec.2016.03.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2024]
|
32
|
Otgon-Uul Z, Suyama S, Onodera H, Yada T. Optogenetic activation of leptin- and glucose-regulated GABAergic neurons in dorsomedial hypothalamus promotes food intake via inhibitory synaptic transmission to paraventricular nucleus of hypothalamus. Mol Metab 2016; 5:709-715. [PMID: 27656408 PMCID: PMC5021668 DOI: 10.1016/j.molmet.2016.06.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 06/15/2016] [Accepted: 06/16/2016] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE The dorsomedial hypothalamus (DMH) has been considered an orexigenic nucleus, since the DMH lesion reduced food intake and body weight and induced resistance to diet-induced obesity. The DMH expresses feeding regulatory neuropeptides and receptors including neuropeptide Y (NPY), cocaine- and amphetamine-regulated transcript (CART), cholecystokinin (CCK), leptin receptor, and melanocortin 3/4 receptors. However, the principal neurons generating the orexigenic function in the DMH remain to be defined. This study aimed to clarify the role of the DMH GABAergic neurons in feeding regulation by using optogenetics and electrophysiological techniques. METHODS We generated the mice expressing ChRFR-C167A, a bistable chimeric channelrhodopsin, selectively in GABAergic neurons of DMH via locally injected adeno-associated virus 2. Food intake after optogenetic activation of DMH GABAergic neurons was measured. Electrophysiological properties of DMH GABAergic neurons were measured using slice patch clamp. RESULTS Optogenetic activation of DMH GABAergic neurons promoted food intake. Leptin hyperpolarized and lowering glucose depolarized half of DMH GABAergic neurons, suggesting their orexigenic property. Optical activation of axonal terminals of DMH GABAergic neurons at the paraventricular nucleus of hypothalamus (PVN), where anorexigenic neurons are localized, increased inhibitory postsynaptic currents on PVN neurons and promoted food intake. CONCLUSION DMH GABAergic neurons are regulated by metabolic signals leptin and glucose and, once activated, promote food intake via inhibitory synaptic transmission to PVN.
Collapse
Affiliation(s)
- Zesemdorj Otgon-Uul
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi, 320-0498, Japan
| | - Shigetomo Suyama
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi, 320-0498, Japan
| | - Hiroshi Onodera
- Photon Science Center of the University of Tokyo, Department of Electrical Engineering of the University of Tokyo, Tokyo, Japan
| | - Toshihiko Yada
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi, 320-0498, Japan.
| |
Collapse
|
33
|
Bell RL, Hauser S, Rodd ZA, Liang T, Sari Y, McClintick J, Rahman S, Engleman EA. A Genetic Animal Model of Alcoholism for Screening Medications to Treat Addiction. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 126:179-261. [PMID: 27055615 PMCID: PMC4851471 DOI: 10.1016/bs.irn.2016.02.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The purpose of this review is to present up-to-date pharmacological, genetic, and behavioral findings from the alcohol-preferring P rat and summarize similar past work. Behaviorally, the focus will be on how the P rat meets criteria put forth for a valid animal model of alcoholism with a highlight on its use as an animal model of polysubstance abuse, including alcohol, nicotine, and psychostimulants. Pharmacologically and genetically, the focus will be on the neurotransmitter and neuropeptide systems that have received the most attention: cholinergic, dopaminergic, GABAergic, glutamatergic, serotonergic, noradrenergic, corticotrophin releasing hormone, opioid, and neuropeptide Y. Herein, we sought to place the P rat's behavioral and neurochemical phenotypes, and to some extent its genotype, in the context of the clinical literature. After reviewing the findings thus far, this chapter discusses future directions for expanding the use of this genetic animal model of alcoholism to identify molecular targets for treating drug addiction in general.
Collapse
Affiliation(s)
- R L Bell
- Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, United States.
| | - S Hauser
- Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Z A Rodd
- Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - T Liang
- Indiana University School of Medicine, Indianapolis, IN, United States
| | - Y Sari
- University of Toledo, Toledo, OH, United States
| | - J McClintick
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - S Rahman
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD, United States
| | - E A Engleman
- Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
34
|
Steyn FJ, Tolle V, Chen C, Epelbaum J. Neuroendocrine Regulation of Growth Hormone Secretion. Compr Physiol 2016; 6:687-735. [PMID: 27065166 DOI: 10.1002/cphy.c150002] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
This article reviews the main findings that emerged in the intervening years since the previous volume on hormonal control of growth in the section on the endocrine system of the Handbook of Physiology concerning the intra- and extrahypothalamic neuronal networks connecting growth hormone releasing hormone (GHRH) and somatostatin hypophysiotropic neurons and the integration between regulators of food intake/metabolism and GH release. Among these findings, the discovery of ghrelin still raises many unanswered questions. One important event was the application of deconvolution analysis to the pulsatile patterns of GH secretion in different mammalian species, including Man, according to gender, hormonal environment and ageing. Concerning this last phenomenon, a great body of evidence now supports the role of an attenuation of the GHRH/GH/Insulin-like growth factor-1 (IGF-1) axis in the control of mammalian aging.
Collapse
Affiliation(s)
- Frederik J Steyn
- University of Queensland Centre for Clinical Research and the School of Biomedical Sciences, University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Virginie Tolle
- Unité Mixte de Recherche en Santé 894 INSERM, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Jacques Epelbaum
- University of Queensland Centre for Clinical Research and the School of Biomedical Sciences, University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| |
Collapse
|
35
|
Tasan RO, Verma D, Wood J, Lach G, Hörmer B, de Lima TCM, Herzog H, Sperk G. The role of Neuropeptide Y in fear conditioning and extinction. Neuropeptides 2016; 55:111-26. [PMID: 26444585 DOI: 10.1016/j.npep.2015.09.007] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 09/10/2015] [Accepted: 09/10/2015] [Indexed: 12/23/2022]
Abstract
While anxiety disorders are the brain disorders with the highest prevalence and constitute a major burden for society, a considerable number of affected people are still treated insufficiently. Thus, in an attempt to identify potential new anxiolytic drug targets, neuropeptides have gained considerable attention in recent years. Compared to classical neurotransmitters they often have a regionally restricted distribution and may bind to several distinct receptor subtypes. Neuropeptide Y (NPY) is a highly conserved neuropeptide that is specifically concentrated in limbic brain areas and signals via at least 5 different G-protein-coupled receptors. It is involved in a variety of physiological processes including the modulation of emotional-affective behaviors. An anxiolytic and stress-reducing property of NPY is supported by many preclinical studies. Whether NPY may also interact with processing of learned fear and fear extinction is comparatively unknown. However, this has considerable relevance since pathological, inappropriate and generalized fear expression and impaired fear extinction are hallmarks of human post-traumatic stress disorder and a major reason for its treatment-resistance. Recent evidence from different laboratories emphasizes a fear-reducing role of NPY, predominantly mediated by exogenous NPY acting on Y1 receptors. Since a reduction of fear expression was also observed in Y1 receptor knockout mice, other Y receptors may be equally important. By acting on Y2 receptors, NPY promotes fear extinction and generates a long-term suppression of fear, two important preconditions that could support cognitive behavioral therapies in human patients. A similar effect has been demonstrated for the closely related pancreatic polypeptide (PP) when acting on Y4 receptors. Preliminary evidence suggests that NPY modulates fear in particular by activation of Y1 and Y2 receptors in the basolateral and central amygdala, respectively. In the basolateral amygdala, NPY signaling activates inhibitory G protein-coupled inwardly-rectifying potassium channels or suppresses hyperpolarization-induced I(h) currents in a Y1 receptor-dependent fashion, favoring a general suppression of neuronal activity. A more complex situation has been described for the central extended amygdala, where NPY reduces the frequency of inhibitory and excitatory postsynaptic currents. In particular the inhibition of long-range central amygdala output neurons may result in a Y2 receptor-dependent suppression of fear. The role of NPY in processes of learned fear and fear extinction is, however, only beginning to emerge, and multiple questions regarding the relevance of endogenous NPY and different receptor subtypes remain elusive. Y2 receptors may be of particular interest for future studies, since they are the most prominent Y receptor subtype in the human brain and thus among the most promising therapeutic drug targets when translating preclinical evidence to potential new therapies for human anxiety disorders.
Collapse
Affiliation(s)
- R O Tasan
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria.
| | - D Verma
- Institute of Physiology I, University of Münster, D-48149 Münster, Germany
| | - J Wood
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - G Lach
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria; Capes Foundation, Ministry of Education of Brazil, 70040-020 Brasília/DF, Brazil
| | - B Hörmer
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - T C M de Lima
- Department of Pharmacology, Federal University of Santa Catarina, 88049-970 Florianópolis, Brazil
| | - H Herzog
- Neuroscience Research Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW 2010, Australia
| | - G Sperk
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
36
|
An Indirect Action Contributes to C-Fos Induction in Paraventricular Hypothalamic Nucleus by Neuropeptide Y. Sci Rep 2016; 6:19980. [PMID: 26813148 PMCID: PMC4728490 DOI: 10.1038/srep19980] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 12/17/2015] [Indexed: 12/03/2022] Open
Abstract
Neuropeptide Y (NPY) is a well-established orexigenic peptide and hypothalamic paraventricular nucleus (PVH) is one major brain site that mediates the orexigenic action of NPY. NPY induces abundant expression of C-Fos, an indicator for neuronal activation, in the PVH, which has been used extensively to examine the underlying NPY orexigenic neural pathways. However, PVH C-Fos induction is in discordance with the abundant expression of NPY receptors, a group of inhibitory Gi protein coupled receptors in the PVH, and with the overall role of PVH neurons in feeding inhibition, suggesting a mechanism of indirect action. Here we showed that the ability of NPY on C-Fos induction in the PVH was blunted in conditions of insulin deficiency and fasting, a condition associated with a high level of NPY and a low level of insulin. Moreover, insulin insufficiency blunted C-Fos induction in the PVH by fasting-induced re-feeding, and insulin and NPY induced c-Fos induction in the same group of PVH neurons. Finally, NPY produced normal C-Fos induction in the PVH with disruption of GABA-A receptors. Thus, our results revealed that PVH C-Fos induction by NPY is mediated by an indirect action, which is at least partially mediated by insulin action, but not GABA-A receptors.
Collapse
|
37
|
Hallberg M. Neuropeptides: metabolism to bioactive fragments and the pharmacology of their receptors. Med Res Rev 2015; 35:464-519. [PMID: 24894913 DOI: 10.1002/med.21323] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
The proteolytic processing of neuropeptides has an important regulatory function and the peptide fragments resulting from the enzymatic degradation often exert essential physiological roles. The proteolytic processing generates, not only biologically inactive fragments, but also bioactive fragments that modulate or even counteract the response of their parent peptides. Frequently, these peptide fragments interact with receptors that are not recognized by the parent peptides. This review discusses tachykinins, opioid peptides, angiotensins, bradykinins, and neuropeptide Y that are present in the central nervous system and their processing to bioactive degradation products. These well-known neuropeptide systems have been selected since they provide illustrative examples that proteolytic degradation of parent peptides can lead to bioactive metabolites with different biological activities as compared to their parent peptides. For example, substance P, dynorphin A, angiotensin I and II, bradykinin, and neuropeptide Y are all degraded to bioactive fragments with pharmacological profiles that differ considerably from those of the parent peptides. The review discusses a selection of the large number of drug-like molecules that act as agonists or antagonists at receptors of neuropeptides. It focuses in particular on the efforts to identify selective drug-like agonists and antagonists mimicking the effects of the endogenous peptide fragments formed. As exemplified in this review, many common neuropeptides are degraded to a variety of smaller fragments but many of the fragments generated have not yet been examined in detail with regard to their potential biological activities. Since these bioactive fragments contain a small number of amino acid residues, they provide an ideal starting point for the development of drug-like substances with ability to mimic the effects of the degradation products. Thus, these substances could provide a rich source of new pharmaceuticals. However, as discussed herein relatively few examples have so far been disclosed of successful attempts to create bioavailable, drug-like agonists or antagonists, starting from the structure of endogenous peptide fragments and applying procedures relying on stepwise manipulations and simplifications of the peptide structures.
Collapse
Affiliation(s)
- Mathias Hallberg
- Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence, Uppsala University, Biomedical Center, Uppsala, Sweden
| |
Collapse
|
38
|
Actions of NPY, and its Y1 and Y2 receptors on pulsatile growth hormone secretion during the fed and fasted state. J Neurosci 2015; 34:16309-19. [PMID: 25471570 DOI: 10.1523/jneurosci.4622-13.2014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The hypothalamic NPY system plays an important role in regulating food intake and energy expenditure. Different biological actions of NPY are assigned to NPY receptor subtypes. Recent studies demonstrated a close relationship between food intake and growth hormone (GH) secretion; however, the mechanism through which endogenous NPY modulates GH release remains unknown. Moreover, conclusive evidence demonstrating a role for NPY and Y-receptors in regulating the endogenous pulsatile release of GH does not exist. We used genetically modified mice (germline Npy, Y1, and Y2 receptor knock-out mice) to assess pulsatile GH secretion under both fed and fasting conditions. Deletion of NPY did not impact fed GH release; however, it reversed the fasting-induced suppression of pulsatile GH secretion. The recovery of GH secretion was associated with a reduction in hypothalamic somatotropin release inhibiting factor (Srif; somatostatin) mRNA expression. Moreover, observations revealed a differential role for Y1 and Y2 receptors, wherein the postsynaptic Y1 receptor suppresses GH secretion in fasting. In contrast, the presynaptic Y2 receptor maintains normal GH output under long-term ad libitum-fed conditions. These data demonstrate an integrated neural circuit that modulates GH release relative to food intake, and provide essential information to address the differential roles of Y1 and Y2 receptors in regulating the release of GH under fed and fasting states.
Collapse
|
39
|
Enman NM, Sabban EL, McGonigle P, Van Bockstaele EJ. Targeting the Neuropeptide Y System in Stress-related Psychiatric Disorders. Neurobiol Stress 2015; 1:33-43. [PMID: 25506604 PMCID: PMC4260418 DOI: 10.1016/j.ynstr.2014.09.007] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Repeated, extreme, or traumatic stressors can elicit pathological effects leading to many negative physical and psychological outcomes. Stressors can precipitate the onset of psychiatric diseases, or exacerbate pre-existing disorders including various anxiety and mood disorders. As stressors can negatively impact human psychiatric health, it is essential to identify neurochemicals that may confer protection from the negative sequelae of repeated or extreme stress exposure. Elucidating the neurobiological underpinnings of stress resilience will enhance our ability to promote resilience to, or recovery from, stress-related psychiatric disease. Herein, we will review the evidence for neuropeptide Y as an endogenous mediator of resilience and its potential relevance for the treatment of stress-related psychiatric diseases. Overview of neuropeptide Y and receptor subtypes in the central nervous system. Alterations of neuropeptide Y in human stress-related psychiatric disorders. Evidence for neuropeptide Y in resilience to stress-related emotionality in rodent behavioral models. Pharmacotherapeutic implications for neuropeptide Y in the treatment of stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Nicole M Enman
- Department of Pharmacology and Physiology, Drexel University, Philadelphia, PA, 19102, USA
| | - Esther L Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, 10595, USA
| | - Paul McGonigle
- Department of Pharmacology and Physiology, Drexel University, Philadelphia, PA, 19102, USA
| | | |
Collapse
|
40
|
Pérez-Fernández J, Megías M, Pombal MA. Cloning, phylogeny, and regional expression of a Y5 receptor mRNA in the brain of the sea lamprey (Petromyzon marinus). J Comp Neurol 2014; 522:1132-54. [PMID: 24127055 DOI: 10.1002/cne.23481] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 09/27/2013] [Accepted: 09/27/2013] [Indexed: 12/12/2022]
Abstract
The NPY receptors known as Y receptors are classified into three subfamilies, Y1, Y2, and Y5, and are involved in different physiological functions. The Y5 receptor is the only member of the Y5 subfamily, and it is present in all vertebrate groups, except for teleosts. Both molecular and pharmacological studies show that Y5 receptor is highly conserved during vertebrate evolution. Furthermore, this receptor is widely expressed in the mammalian brain, including the hypothalamus, where it is thought to take part in feeding and homeostasis regulation. Lampreys belong to the agnathan lineage, and they are thought to have branched out between the two whole-genome duplications that occurred in vertebrates. Therefore, they are in a key position for studies on the evolution of gene families in vertebrates. Here we report the cloning, phylogeny, and brain expression pattern of the sea lamprey Y5 receptor. In phylogenetic studies, the lamprey Y5 receptor clusters in a basal position, together with Y5 receptors of other vertebrates. The mRNA of this receptor is broadly expressed in the lamprey brain, being especially abundant in hypothalamic areas. Its expression pattern is roughly similar to that reported for other vertebrates and parallels the expression pattern of the Y1 receptor subtype previously described by our group, as it occurs in mammals. Altogether, these results confirm that a Y5 receptor is present in lampreys, thus being highly conserved during the evolution of vertebrates, and suggest that it is involved in many brain functions, the only known exception being teleosts.
Collapse
Affiliation(s)
- Juan Pérez-Fernández
- Neurolam Group, Department of Functional Biology and Health Sciences, Faculty of Biology, University of Vigo, 36310-Vigo, Spain
| | | | | |
Collapse
|
41
|
Cassaglia PA, Shi Z, Li B, Reis WL, Clute-Reinig NM, Stern JE, Brooks VL. Neuropeptide Y acts in the paraventricular nucleus to suppress sympathetic nerve activity and its baroreflex regulation. J Physiol 2014; 592:1655-75. [PMID: 24535439 DOI: 10.1113/jphysiol.2013.268763] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Neuropeptide Y (NPY), a brain neuromodulator that has been strongly implicated in the regulation of energy balance, also acts centrally to inhibit sympathetic nerve activity (SNA); however, the site and mechanism of action are unknown. In chloralose-anaesthetized female rats, nanoinjection of NPY into the paraventricular nucleus of the hypothalamus (PVN) dose-dependently suppressed lumbar SNA (LSNA) and its baroreflex regulation, and these effects were blocked by prior inhibition of NPY Y1 or Y5 receptors. Moreover, PVN injection of Y1 and Y5 receptor antagonists in otherwise untreated rats increased basal and baroreflex control of LSNA, indicating that endogenous NPY tonically inhibits PVN presympathetic neurons. The sympathoexcitation following blockade of PVN NPY inhibition was eliminated by prior PVN nanoinjection of the melanocortin 3/4 receptor inhibitor SHU9119. Moreover, presympathetic neurons, identified immunohistochemically using cholera toxin b neuronal tract tracing from the rostral ventrolateral medulla (RVLM), express NPY Y1 receptor immunoreactivity, and patch-clamp recordings revealed that both NPY and α-melanocyte-stimulating hormone (α-MSH) inhibit and stimulate, respectively, PVN-RVLM neurons. Collectively, these data suggest that PVN NPY inputs converge with α-MSH to influence presympathetic neurons. Together these results identify endogenous NPY as a novel and potent inhibitory neuromodulator within the PVN that may contribute to changes in SNA that occur in states associated with altered energy balance, such as obesity and pregnancy.
Collapse
Affiliation(s)
- Priscila A Cassaglia
- Oregon Health & Science University, Department of Physiology and Pharmacology, 1381 SW Sam Jackson Park Rd - L334, Portland, OR 97239, USA.
| | | | | | | | | | | | | |
Collapse
|
42
|
Shi YC, Lin Z, Lau J, Zhang H, Yagi M, Kanzler I, Sainsbury A, Herzog H, Lin S. PYY3-36 and pancreatic polypeptide reduce food intake in an additive manner via distinct hypothalamic dependent pathways in mice. Obesity (Silver Spring) 2013; 21:E669-78. [PMID: 23804428 DOI: 10.1002/oby.20534] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 05/18/2013] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Peptide YY (PYY3-36) and pancreatic polypeptide (PP) potently inhibit food intake in rodents and humans, however, it is unclear whether they have any synergistic/additive interaction in decreasing food intake. DESIGN AND METHODS Fasted WT, Y2(-) (/) (-) , Y4(-) (/) (-) , or Y2Y4(-) (/) (-) mice were i.p. administrated with saline, PYY3-36, and/or PP. RESULTS Combined injection of PYY3-36 and PP reduces food intake in an additive manner was demonstrated in this study. This effect is mediated via Y2 and Y4 receptors, respectively. It was demonstrated that PYY3-36 and PP activate distinct neuronal pathways in the hypothalamus, as demonstrated by immunostaining for c-fos, which shows distinct patterns in response to either hormone. After PYY3-36 injection, neurons in the dorsal aspect of the arcuate nucleus (Arc), paraventricular nucleus, and dorso-medial nucleus of the hypothalamus (DMH) are activated with minimal responses seen in the ventro-medial nucleus of the hypothalamus (VMH) and lateral hypothalamic area (LHA) of WT mice. These effects are absent in Y2(-) (/) (-) mice. PP activates preferably the lateral aspect of the Arc, the DMH, VMH, and LHA in a Y4 receptor-dependent manner. Importantly, the expression pattern of c-fos immunoreactive neurons induced by combined treatment appears to be the sum of the effects of single treatments rather than a result of synergistic interaction. CONCLUSIONS These findings demonstrate that PYY3-36 and PP activate distinct pathways in the hypothalamus to reduce food intake in an additive manner.
Collapse
Affiliation(s)
- Yan-Chuan Shi
- Neuroscience Research Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia; Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Casillas-Espinosa PM, Powell KL, O'Brien TJ. Regulators of synaptic transmission: roles in the pathogenesis and treatment of epilepsy. Epilepsia 2013; 53 Suppl 9:41-58. [PMID: 23216578 DOI: 10.1111/epi.12034] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Synaptic transmission is the communication between a presynaptic and a postsynaptic neuron, and the subsequent processing of the signal. These processes are complex and highly regulated, reflecting their importance in normal brain functioning and homeostasis. Sustaining synaptic transmission depends on the continuing cycle of synaptic vesicle formation, release, and endocytosis, which requires proteins such as dynamin, syndapin, synapsin, and synaptic vesicle protein 2A. Synaptic transmission is regulated by diverse mechanisms, including presynaptic modulators of synaptic vesicle formation and release, postsynaptic receptors and signaling, and modulators of neurotransmission. Neurotransmitters released presynaptically can bind to their postsynaptic receptors, the inhibitory γ-aminobutyric acid (GABA)ergic receptors or the excitatory glutamate receptors. Once released, glutamate activates a variety of postsynaptic receptors including α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), N-methyl-D-aspartate (NMDA), kainate, and metabotropic receptors. The activation of the receptors triggers downstream signaling cascades generating a vast array of effects, which can be modulated by a numerous auxiliary regulatory subunits. Moreover, different neuropeptides such as neuropeptide Y, brain-derived neurotrophic factor (BDNF), somatostatin, ghrelin, and galanin, act as regulators of diverse synaptic functions and along with the classic neurotransmitters. Abnormalities in the regulation of synaptic transmission play a critical role in the pathogenesis of numerous brain diseases, including epilepsy. This review focuses on the different mechanisms involved in the regulation of synaptic transmission, which may play a role in the pathogenesis of epilepsy: the presynaptic modulators of synaptic vesicle formation and release, postsynaptic receptors, and modulators of neurotransmission, including the mechanism by which drugs can modulate the frequency and severity of epileptic seizures.
Collapse
Affiliation(s)
- Pablo M Casillas-Espinosa
- The Departments of Medicine and Neurology, The Royal Melbourne Hospital, The Melbourne Brain Centre, The University of Melbourne, Parkville, Victoria, Australia
| | | | | |
Collapse
|
44
|
Pérez-Fernández J, Megías M, Pombal MA. Distribution of a Y1 receptor mRNA in the brain of two Lamprey species, the sea lamprey (Petromyzon marinus) and the river Lamprey (Lampetra fluviatilis). J Comp Neurol 2013; 521:426-47. [PMID: 22740099 DOI: 10.1002/cne.23180] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 05/23/2012] [Accepted: 06/22/2012] [Indexed: 11/09/2022]
Abstract
The neuropeptide Y system consists of several neuropeptides acting through a broad number of receptor subtypes, the NPY family of receptors. NPY receptors are divided into three subfamilies (Y1, Y2, and Y5) that display a complex evolutionary history due to local and large-scale gene duplication events and gene losses. Lampreys emerged from a basal branch of the tree of vertebrates and they are in a key position to shed light on the evolutionary history of the NPY system. One member of the Y1 subfamily has been reported in agnathans, but the phylogenetic tree of the Y1 subfamily is not yet clear. We cloned the sequences of the Y1-subtype receptor of Petromyzon marinus and Lampetra fluviatilis to study the expression pattern of this receptor in lampreys by in situ hybridization and to analyze the phylogeny of the Y1-subfamily receptors in vertebrates. The phylogenetic study showed that the Y1 receptor of lampreys is basal to the Y1/6 branch of the Y1-subfamily receptors. In situ hybridization showed that the Y1 receptor is widely expressed throughout the brain of lampreys, with some regions showing numerous positive neurons, as well as the presence of numerous cerebrospinal fluid-contacting cells in the spinal cord. This broad distribution of the lamprey Y1 receptor is more similar to that found in other vertebrates for the Y1 receptor than that of the other members of the Y1 subfamily: Y4, Y8, and Y6 receptors. Both phylogenetic relationship and expression pattern suggest that this receptor is a Y1 receptor.
Collapse
Affiliation(s)
- Juan Pérez-Fernández
- Neurolam Group, Department of Functional Biology and Health Sciences, University of Vigo, Vigo, Spain
| | | | | |
Collapse
|
45
|
Rojas JM, Stafford JM, Saadat S, Printz RL, Beck-Sickinger AG, Niswender KD. Central nervous system neuropeptide Y signaling via the Y1 receptor partially dissociates feeding behavior from lipoprotein metabolism in lean rats. Am J Physiol Endocrinol Metab 2012; 303:E1479-88. [PMID: 23074243 PMCID: PMC3532466 DOI: 10.1152/ajpendo.00351.2012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Elevated plasma triglyceride (TG) levels contribute to an atherogenic dyslipidemia that is associated with obesity, diabetes, and metabolic syndrome. Numerous models of obesity are characterized by increased central nervous system (CNS) neuropeptide Y (NPY) tone that contributes to excess food intake and obesity. Previously, we demonstrated that intracerebroventricular (icv) administration of NPY in lean fasted rats also elevates hepatic production of very low-density lipoprotein (VLDL)-TG. Thus, we hypothesize that elevated CNS NPY action contributes to not only the pathogenesis of obesity but also dyslipidemia. Here, we sought to determine whether the effects of NPY on feeding and/or obesity are dissociable from effects on hepatic VLDL-TG secretion. Pair-fed, icv NPY-treated, chow-fed Long-Evans rats develop hypertriglyceridemia in the absence of increased food intake and body fat accumulation compared with vehicle-treated controls. We then modulated CNS NPY signaling by icv injection of selective NPY receptor agonists and found that Y1, Y2, Y4, and Y5 receptor agonists all induced hyperphagia in lean, ad libitum chow-fed Long-Evans rats, with the Y2 receptor agonist having the most pronounced effect. Next, we found that at equipotent doses for food intake NPY Y1 receptor agonist had the most robust effect on VLDL-TG secretion, a Y2 receptor agonist had a modest effect, and no effect was observed for Y4 and Y5 receptor agonists. These findings, using selective agonists, suggest the possibility that the effect of CNS NPY signaling on hepatic VLDL-TG secretion may be relatively dissociable from effects on feeding behavior via the Y1 receptor.
Collapse
|
46
|
Holzer P, Reichmann F, Farzi A. Neuropeptide Y, peptide YY and pancreatic polypeptide in the gut-brain axis. Neuropeptides 2012; 46:261-74. [PMID: 22979996 PMCID: PMC3516703 DOI: 10.1016/j.npep.2012.08.005] [Citation(s) in RCA: 330] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 08/04/2012] [Accepted: 08/09/2012] [Indexed: 02/06/2023]
Abstract
The gut-brain axis refers to the bidirectional communication between the gut and the brain. Four information carriers (vagal and spinal afferent neurons, immune mediators such as cytokines, gut hormones and gut microbiota-derived signalling molecules) transmit information from the gut to the brain, while autonomic neurons and neuroendocrine factors carry outputs from the brain to the gut. The members of the neuropeptide Y (NPY) family of biologically active peptides, NPY, peptide YY (PYY) and pancreatic polypeptide (PP), are expressed by cell systems at distinct levels of the gut-brain axis. PYY and PP are exclusively expressed by endocrine cells of the digestive system, whereas NPY is found at all levels of the gut-brain and brain-gut axis. The major systems expressing NPY comprise enteric neurons, primary afferent neurons, several neuronal pathways throughout the brain and sympathetic neurons. In the digestive tract, NPY and PYY inhibit gastrointestinal motility and electrolyte secretion and in this way modify the input to the brain. PYY is also influenced by the intestinal microbiota, and NPY exerts, via stimulation of Y1 receptors, a proinflammatory action. Furthermore, the NPY system protects against distinct behavioural disturbances caused by peripheral immune challenge, ameliorating the acute sickness response and preventing long-term depression. At the level of the afferent system, NPY inhibits nociceptive input from the periphery to the spinal cord and brainstem. In the brain, NPY and its receptors (Y1, Y2, Y4, Y5) play important roles in regulating food intake, energy homeostasis, anxiety, mood and stress resilience. In addition, PP and PYY signal to the brain to attenuate food intake, anxiety and depression-related behaviour. These findings underscore the important role of the NPY-Y receptor system at several levels of the gut-brain axis in which NPY, PYY and PP operate both as neural and endocrine messengers.
Collapse
Affiliation(s)
- Peter Holzer
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, A-8010 Graz, Austria.
| | | | | |
Collapse
|
47
|
Garcia FD, Coquerel Q, do Rego JC, Cravezic A, Bole-Feysot C, Kiive E, Déchelotte P, Harro J, Fetissov SO. Anti-neuropeptide Y plasma immunoglobulins in relation to mood and appetite in depressive disorder. Psychoneuroendocrinology 2012; 37:1457-67. [PMID: 22365482 DOI: 10.1016/j.psyneuen.2012.01.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Revised: 01/31/2012] [Accepted: 01/31/2012] [Indexed: 12/27/2022]
Abstract
Depression and eating disorders are frequently associated, but the molecular pathways responsible for co-occurrence of altered mood, appetite and body weight are not yet fully understood. Neuropeptide Y (NPY) has potent antidepressant and orexigenic properties and low central NPY levels have been reported in major depression. In the present study, we hypothesized that in patients with major depression alteration of mood, appetite and body weight may be related to NPY-reactive autoantibodies (autoAbs). To test this hypothesis, we compared plasma levels and affinities of NPY-reactive autoAbs between patients with major depression and healthy controls. Then, to evaluate if changes of NPY autoAb properties can be causally related to altered mood and appetite, we developed central and peripheral passive transfer models of human autoAbs in mice and studied depressive-like behavior in forced-swim test and food intake. We found that plasma levels of NPY IgG autoAbs were lower in patients with moderate but not with mild depression correlating negatively with the Montgomery-Åsberg Depression Rating Scale scores and with immobility time of the forced-swim test in mice after peripheral injection of autoAbs. No significant differences in NPY IgG autoAb affinities between patients with depression and controls were found, but higher affinity of IgG autoAbs for NPY was associated with lower body mass index and prevented NPY-induced orexigenic response in mice after their central injection. These data suggest that changes of plasma levels of anti-NPY autoAbs are relevant to altered mood, while changes of their affinity may participate in altered appetite and body weight in patients with depressive disorder.
Collapse
Affiliation(s)
- Frederico D Garcia
- Nutrition, Gut and Brain Laboratory, Inserm U1073, Institute for Research and Innovation in Biomedicine-IRIB, Rouen University, Normandy 76183, France
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Reaux-Le Goazigo A, Bodineau L, De Mota N, Jeandel L, Chartrel N, Knauf C, Raad C, Valet P, Llorens-Cortes C. Apelin and the proopiomelanocortin system: a new regulatory pathway of hypothalamic α-MSH release. Am J Physiol Endocrinol Metab 2011; 301:E955-66. [PMID: 21846903 DOI: 10.1152/ajpendo.00090.2011] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neuronal networks originating in the hypothalamic arcuate nucleus (Arc) play a fundamental role in controlling energy balance. In the Arc, neuropeptide Y (NPY)-producing neurons stimulate food intake, whereas neurons releasing the proopiomelanocortin (POMC)-derived peptide α-melanocyte-stimulating hormone (α-MSH) strongly decrease food intake. There is growing evidence to suggest that apelin and its receptor may play a role in the central control of food intake, and both are concentrated in the Arc. We investigated the presence of apelin and its receptor in Arc NPY- and POMC-containing neurons and the effects of apelin on α-MSH release in the hypothalamus. We showed, by immunofluorescence and confocal microscopy, that apelin-immunoreactive (IR) neuronal cell bodies were distributed throughout the rostrocaudal extent of the Arc and that apelin was strongly colocalized with POMC, but weakly colocalized with NPY. However, there were numerous NPY-IR nerve fibers close to the apelin-IR neuronal cell bodies. By combining in situ hybridization with immunohistochemistry, we demonstrated the presence of apelin receptor mRNA in Arc POMC neurons. Moreover, using a perifusion technique for hypothalamic explants, we demonstrated that apelin-17 (K17F) increased α-MSH release, suggesting that apelin released somato-dendritically or axonally from POMC neurons may stimulate α-MSH release in an autocrine manner. Consistent with these data, hypothalamic apelin levels were found to be higher in obese db/db mice and fa/fa Zucker rats than in wild-type animals. These findings support the hypothesis that central apelin is involved in regulating body weight and feeding behavior through the direct stimulation of α-MSH release.
Collapse
Affiliation(s)
- Annabelle Reaux-Le Goazigo
- Institut National de Santé et de Recherche Médicale, Unité Mixte de Recherche S 691, Centre for Interdisciplinary Research in Biology, Collège de France, and Université Pierre et Marie Curie-Paris 6, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
The neuropeptide Y system: Pathophysiological and therapeutic implications in obesity and cancer. Pharmacol Ther 2011; 131:91-113. [DOI: 10.1016/j.pharmthera.2011.03.011] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2011] [Accepted: 03/07/2011] [Indexed: 12/28/2022]
|
50
|
Griffith DA, Hargrove DM, Maurer TS, Blum CA, De Lombaert S, Inthavongsay JK, Klade LE, Mack CM, Rose CR, Sanders MJ, Carpino PA. Discovery and evaluation of pyrazolo[1,5-a]pyrimidines as neuropeptide Y1 receptor antagonists. Bioorg Med Chem Lett 2011; 21:2641-5. [DOI: 10.1016/j.bmcl.2010.12.116] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Revised: 12/18/2010] [Accepted: 12/21/2010] [Indexed: 11/26/2022]
|