1
|
Korkmaz OT, Saydam F, Dalkiran B, Değirmenci İ, Tunçel N. Vasoactive Intestinal Peptide (VIP) and its Receptors in Adipose Tissue: Implications for Cold Stress Adaptation. Cell Biochem Biophys 2024:10.1007/s12013-024-01606-0. [PMID: 39550744 DOI: 10.1007/s12013-024-01606-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2024] [Indexed: 11/18/2024]
Abstract
Adipose tissue represents an organ that is highly dynamic and contributes toward vital survival events such as immune responses, lactation, metabolism fuel, and thermogenesis. Data emerging from recent studies support the notion of adipose tissue being organized into a complex system characterized by a discrete anatomy, elevated physiological plasticity, and specific vascular and nerve supplies. Vasoactive intestinal peptide (VIP), along with its receptors, type 1 (VPAC1) and type 2 (VPAC2), has been implicated in various physiological and pathophysiological processes. However, studies on VIP and its receptors in adipose tissue are limited. To explore VIP's presence and activity, as well as its adipose tissue-based receptors, we conducted a study on isolated adipocytes and adipose tissue from inguinal white adipose tissue (WAT) and interscapular brown adipose tissue (BAT) in normal and cold-stressed rats. Our findings indicate the presence of the gene expression VIP and VPAC1 in both WAT and BAT under normal conditions, while VPAC2 was absent. In both WAT and BAT, cold exposure upregulated VIP gene expression. However, the response of VIP receptors to cold exposure is controversial. VPAC2 gene expression was induced in both WAT and BAT, while VPAC1 gene expression presented no change of significance in BAT and a slight reduction in WAT. Additionally, VIP, VPAC1, and VPAC2 proteins were identified from Western blot studies on white and brown adipocytes. After exposure to cold there was an increase of significance in the VIP, VPAC1, and VPAC2 protein levels. This study provides novel insights into how VIP and its receptors alter gene expression and protein levels in adipose tissue and adipocytes during cold stress, indicating their potential involvement in adipose tissue regulation. The findings propose VIP's potentially crucial role in adipose tissue's adaptation to cold stress by affecting the metabolic and biochemical functions of subcutaneous and interscapular adipocytes, with potentially significant implications in the context of developing therapies targeting metabolic disorders.
Collapse
Affiliation(s)
- Orhan Tansel Korkmaz
- Department of Physiology, Faculty of Medicine, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey.
| | - Faruk Saydam
- Department of Medical Biology, Medical Faculty, Recep Tayyip Erdogan University, 53100, Rize, Turkey
| | - Bahar Dalkiran
- Department of Physiology, Faculty of Medicine, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
| | - İrfan Değirmenci
- Department of Medical Biology, Medical Faculty, Kutahya Health Sciences University, 43020, Kütahya, Turkey
| | - Neşe Tunçel
- Department of Physiology, Faculty of Medicine, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
| |
Collapse
|
2
|
Sayers S, Le N, Wagner EJ. The role of pituitary adenylate cyclase-activating polypeptide neurons in the hypothalamic ventromedial nucleus and the cognate PAC1 receptor in the regulation of hedonic feeding. Front Nutr 2024; 11:1437526. [PMID: 39234295 PMCID: PMC11371718 DOI: 10.3389/fnut.2024.1437526] [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: 05/23/2024] [Accepted: 07/29/2024] [Indexed: 09/06/2024] Open
Abstract
Obesity is a health malady that affects mental, physical, and social health. Pathology includes chronic imbalance between energy intake and expenditure, likely facilitated by dysregulation of the mesolimbic dopamine (DA) pathway. We explored the role of pituitary adenylate cyclase-activating polypeptide (PACAP) neurons in the hypothalamic ventromedial nucleus (VMN) and the PACAP-selective (PAC1) receptor in regulating hedonic feeding. We hypothesized that VMN PACAP neurons would inhibit reward-encoding mesolimbic (A10) dopamine neurons via PAC1 receptor activation and thereby suppress impulsive consumption brought on by intermittent exposure to highly palatable food. Visualized whole-cell patch clamp recordings coupled with in vivo behavioral experiments were utilized in wildtype, PACAP-cre, TH-cre, and TH-cre/PAC1 receptor-floxed mice. We found that bath application of PACAP directly inhibited preidentified A10 dopamine neurons in the ventral tegmental area (VTA) from TH-cre mice. This inhibitory action was abrogated by the selective knockdown of the PAC1 receptor in A10 dopamine neurons. PACAP delivered directly into the VTA decreases binge feeding accompanied by reduced meal size and duration in TH-cre mice. These effects are negated by PAC1 receptor knockdown in A10 dopamine neurons. Additionally, apoptotic ablation of VMN PACAP neurons increased binge consumption in both lean and obese, male and female PACAP-cre mice relative to wildtype controls. These findings demonstrate that VMN PACAP neurons blunt impulsive, binge feeding behavior by activating PAC1 receptors to inhibit A10 dopamine neurons. As such, they impart impactful insight into potential treatment strategies for conditions such as obesity and food addiction.
Collapse
Affiliation(s)
- Sarah Sayers
- College of Osteopathic Medicine of the Pacific, Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Nikki Le
- College of Osteopathic Medicine of the Pacific, Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Edward J Wagner
- College of Osteopathic Medicine of the Pacific, Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, United States
| |
Collapse
|
3
|
Mata-Pacheco V, Hernandez J, Varma N, Xu J, Sayers S, Le N, Wagner EJ. Dynamic, sex- and diet-specific pleiotropism in the PAC1 receptor-mediated regulation of arcuate proopiomelanocortin and Neuropeptide Y/Agouti related peptide neuronal excitability by anorexigenic ventromedial nucleus PACAP neurons. J Neuroendocrinol 2024; 36:e13357. [PMID: 38056947 DOI: 10.1111/jne.13357] [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: 04/28/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 12/08/2023]
Abstract
This study furthers the investigation of how pituitary adenylate cyclase activating polypeptide (PACAP) and the PAC1 receptor (PAC1R) regulate the homeostatic energy balance circuitry. We hypothesized that apoptotic ablation of PACAP neurones in the hypothalamic ventromedial nucleus (VMN) would affect both energy intake and energy expenditure. We also hypothesized that selective PAC1R knockdown would impair the PACAP-induced excitation in anorexigenic proopiomelanocortin (POMC) neurones and inhibition of orexigenic neuropeptide Y (NPY)/agouti-related peptide (AgRP) neurones in the hypothalamic arcuate nucleus (ARC). The results show CASPASE-3-induced ablation of VMN PACAP neurones leads to increased energy intake and meal frequency as well as decreased energy expenditure in lean animals. The effects were more robust in obese males, whereas we saw the opposite effects in obese females. We then utilized visualized whole-cell patch clamp recordings in hypothalamic slices. PAC1R knockdown in POMC neurones diminishes the PACAP-induced depolarization, increase in firing, decreases in energy intake and meal size, as well as increases in CO2 production and O2 consumption. Similarly, the lack of expression of the PAC1R in NPY/AgRP neurones greatly attenuates the PACAP-induced hyperpolarization, suppression of firing, decreases in energy intake and meal frequency, as well as increases in energy expenditure. The PACAP response in NPY/AgRP neurones switched from predominantly inhibitory to excitatory in fasted animals. Finally, the anorexigenic effect of PACAP was potentiated when oestradiol was injected into the ARC in ovariectomized females. This study demonstrates the critical role of anorexigenic VMN PACAP neurones and the PAC1R in exciting POMC and inhibiting NPY/AgRP neurons to control homeostatic feeding.
Collapse
Affiliation(s)
- Veronica Mata-Pacheco
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, California, USA
| | - Jennifer Hernandez
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Nandini Varma
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Jenny Xu
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Sarah Sayers
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Nikki Le
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Edward J Wagner
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, California, USA
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| |
Collapse
|
4
|
Deem JD, Tingley D, Bjerregaard AM, Secher A, Chan O, Uzo C, Richardson NE, Giering E, Doan T, Phan BA, Wu B, Scarlett JM, Morton GJ, Schwartz MW. Identification of Hypothalamic Glucoregulatory Neurons That Sense and Respond to Changes in Glycemia. Diabetes 2023; 72:1207-1213. [PMID: 37347793 PMCID: PMC10450823 DOI: 10.2337/db23-0139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 06/19/2023] [Indexed: 06/24/2023]
Abstract
To investigate whether glucoregulatory neurons in the hypothalamus can sense and respond to physiological variation in the blood glucose (BG) level, we combined continuous arterial glucose monitoring with continuous measures of the activity of a specific subset of neurons located in the hypothalamic ventromedial nucleus that express pituitary adenylate cyclase activating peptide (VMNPACAP neurons) obtained using fiber photometry. Data were collected in conscious, free-living mice during a 1-h baseline monitoring period and a subsequent 2-h intervention period during which the BG level was raised either by consuming a chow or a high-sucrose meal or by intraperitoneal glucose injection. Cross-correlation analysis revealed that, following a 60- to 90-s delay, interventions that raise the BG level reliably associate with reduced VMNPACAP neuron activity (P < 0.01). In addition, a strong positive correlation between BG and spontaneous VMNPACAP neuron activity was observed under basal conditions but with a much longer (∼25 min) temporal offset, consistent with published evidence that VMNPACAP neuron activation raises the BG level. Together, these findings are suggestive of a closed-loop system whereby VMNPACAP neuron activation increases the BG level; detection of a rising BG level, in turn, feeds back to inhibit these neurons. To our knowledge, these findings constitute the first evidence of a role in glucose homeostasis for glucoregulatory neurocircuits that, like pancreatic β-cells, sense and respond to physiological variation in glycemia. ARTICLE HIGHLIGHTS By combining continuous arterial glucose monitoring with fiber photometry, studies investigated whether neurons in the murine ventromedial nucleus that express pituitary adenylate cyclase activating peptide (VMNPACAP neurons) detect and respond to changes in glycemia in vivo. VMNPACAP neuron activity rapidly decreases (within <2 min) when the blood glucose level is raised by either food consumption or glucose administration. Spontaneous VMNPACAP neuron activity also correlates positively with glycemia, but with a longer temporal offset, consistent with reports that hyperglycemia is induced by experimental activation of these neurons. Like pancreatic β-cells, neurons in the hypothalamic ventromedial nucleus appear to sense and respond to physiological variation in glycemia.
Collapse
Affiliation(s)
- Jennifer D. Deem
- University of Washington Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA
| | - David Tingley
- Beth Israel-Deaconess Medical Center, Harvard University School of Medicine, Boston, MA
| | | | - Anna Secher
- Data Science Intelligence, Global Drug Discovery, Novo Nordisk A/S, Maaloev, Denmark
| | - Owen Chan
- Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Chukwuemeka Uzo
- Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Nicole E. Richardson
- University of Washington Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA
| | - Elizabeth Giering
- University of Washington Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA
- Veterans Affairs Puget Sound Health Care System, Seattle, WA
| | - Tammy Doan
- University of Washington Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA
| | - Bao A. Phan
- University of Washington Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA
| | - Brandon Wu
- University of Washington Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA
| | - Jarrad M. Scarlett
- University of Washington Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA
- Division of Pediatric Gastroenterology and Hepatology, Department of Pediatrics, Seattle Children’s Hospital, Seattle, WA
| | - Gregory J. Morton
- University of Washington Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA
| | - Michael W. Schwartz
- University of Washington Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
5
|
Pandher PK, Rahim Y, Timms KP, Filatov E, Short LI, Gray SL. Reference gene recommendations and PACAP receptor expression in murine sympathetic ganglia of the autonomic nervous system that innervate adipose tissues after chronic cold exposure. J Neuroendocrinol 2023; 35:e13313. [PMID: 37404042 DOI: 10.1111/jne.13313] [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: 11/04/2022] [Revised: 06/01/2023] [Accepted: 06/04/2023] [Indexed: 07/06/2023]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is an important regulator of the stress response in mammals, influencing both the hypothalamic-pituitary-adrenal (HPA) axis and the sympathetic nervous system (SNS). PACAP has been reported to influence energy homeostasis, including adaptive thermogenesis, an energy burning process in adipose tissue regulated by the SNS in response to cold stress and overfeeding. While research suggests PACAP acts centrally at the level of the hypothalamus, knowledge of PACAP's role within the sympathetic nerves innervating adipose tissues in response to metabolic stressors is limited. This work shows, for the first time, gene expression of PACAP receptors in stellate ganglia and highlights some differential expression with housing temperature. Additionally, we present our dissection protocol, analysis of tyrosine hydroxylase gene expression as a molecular biomarker for catecholamine producing tissue and recommend three stable reference genes for the normalization of quantitative real time-polymerase chain reaction (qRT-PCR) data when working with this tissue. This study adds to information about neuropeptide receptor expression in peripheral ganglia of the sympathetic nervous system innervating adipose tissue and provides insight into PACAP's role in the regulation of energy metabolism.
Collapse
Affiliation(s)
- Parleen K Pandher
- Northern Medical Program, Division of Medical Sciences, University of Northern British Columbia, Prince George, British Columbia, Canada
| | - Yamna Rahim
- Northern Medical Program, Division of Medical Sciences, University of Northern British Columbia, Prince George, British Columbia, Canada
| | - Katherine P Timms
- Northern Medical Program, Division of Medical Sciences, University of Northern British Columbia, Prince George, British Columbia, Canada
| | - Ekaterina Filatov
- Northern Medical Program, Division of Medical Sciences, University of Northern British Columbia, Prince George, British Columbia, Canada
| | - Landon I Short
- Northern Medical Program, Division of Medical Sciences, University of Northern British Columbia, Prince George, British Columbia, Canada
| | - Sarah L Gray
- Northern Medical Program, Division of Medical Sciences, University of Northern British Columbia, Prince George, British Columbia, Canada
| |
Collapse
|
6
|
Vu JP, Luong L, Sanford D, Oh S, Kuc A, Pisegna R, Lewis M, Pisegna JR, Germano PM. PACAP and VIP Neuropeptides' and Receptors' Effects on Appetite, Satiety and Metabolism. BIOLOGY 2023; 12:1013. [PMID: 37508442 PMCID: PMC10376325 DOI: 10.3390/biology12071013] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023]
Abstract
The overwhelming increase in the prevalence of obesity and related disorders in recent years is one of the greatest threats to the global healthcare system since it generates immense healthcare costs. As the prevalence of obesity approaches epidemic proportions, the importance of elucidating the mechanisms regulating appetite, satiety, body metabolism, energy balance and adiposity has garnered significant attention. Currently, gastrointestinal (GI) bariatric surgery remains the only approach capable of achieving successful weight loss. Appetite, satiety, feeding behavior, energy intake and expenditure are regulated by central and peripheral neurohormonal mechanisms that have not been fully elucidated yet. Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) and Vasoactive Intestinal Polypeptide (VIP) are members of a family of regulatory peptides that are widely distributed in parallel with their specific receptors, VPAC1R, VPAC2R and PAC1R, in the central nervous system (CNS) and in the periphery, such as in the gastrointestinal tract and its associated organs and immune cells. PACAP and VIP have been reported to play an important role in the regulation of body phenotype, metabolism and homeostatic functions. The purpose of this review is to present recent data on the effects of PACAP, VIP, VPAC1R, VPAC2R and PAC1R on the modulation of appetite, satiety, metabolism, calorie intake and fat accumulation, to evaluate their potential use as therapeutic targets for the treatment of obesity and metabolic syndrome.
Collapse
Affiliation(s)
- John P Vu
- Research Service, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- CURE/Digestive Diseases Research Center, Department of Medicine, University of California, Los Angeles, CA 90073, USA
| | - Leon Luong
- Research Service, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- CURE/Digestive Diseases Research Center, Department of Medicine, University of California, Los Angeles, CA 90073, USA
| | - Daniel Sanford
- Research Service, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- CURE/Digestive Diseases Research Center, Department of Medicine, University of California, Los Angeles, CA 90073, USA
| | - Suwan Oh
- Research Service, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- CURE/Digestive Diseases Research Center, Department of Medicine, University of California, Los Angeles, CA 90073, USA
| | - Alma Kuc
- Research Service, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Rita Pisegna
- Research Service, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Michael Lewis
- Division of Hematology and Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90078, USA
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Pathology, Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, CA 90073, USA
| | - Joseph R Pisegna
- Research Service, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- CURE/Digestive Diseases Research Center, Department of Medicine, University of California, Los Angeles, CA 90073, USA
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System and Department of Medicine, Los Angeles, CA 90073, USA
- Division of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Patrizia M Germano
- Research Service, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- CURE/Digestive Diseases Research Center, Department of Medicine, University of California, Los Angeles, CA 90073, USA
- Division of Pulmonary and Critical Care, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| |
Collapse
|
7
|
Le N, Sayers S, Mata-Pacheco V, Wagner EJ. The PACAP Paradox: Dynamic and Surprisingly Pleiotropic Actions in the Central Regulation of Energy Homeostasis. Front Endocrinol (Lausanne) 2022; 13:877647. [PMID: 35721722 PMCID: PMC9198406 DOI: 10.3389/fendo.2022.877647] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/30/2022] [Indexed: 12/11/2022] Open
Abstract
Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP), a pleiotropic neuropeptide, is widely distributed throughout the body. The abundance of PACAP expression in the central and peripheral nervous systems, and years of accompanying experimental evidence, indicates that PACAP plays crucial roles in diverse biological processes ranging from autonomic regulation to neuroprotection. In addition, PACAP is also abundantly expressed in the hypothalamic areas like the ventromedial and arcuate nuclei (VMN and ARC, respectively), as well as other brain regions such as the nucleus accumbens (NAc), bed nucleus of stria terminalis (BNST), and ventral tegmental area (VTA) - suggesting that PACAP is capable of regulating energy homeostasis via both the homeostatic and hedonic energy balance circuitries. The evidence gathered over the years has increased our appreciation for its function in controlling energy balance. Therefore, this review aims to further probe how the pleiotropic actions of PACAP in regulating energy homeostasis is influenced by sex and dynamic changes in energy status. We start with a general overview of energy homeostasis, and then introduce the integral components of the homeostatic and hedonic energy balance circuitries. Next, we discuss sex differences inherent to the regulation of energy homeostasis via these two circuitries, as well as the activational effects of sex steroid hormones that bring about these intrinsic disparities between males and females. Finally, we explore the multifaceted role of PACAP in regulating homeostatic and hedonic feeding through its actions in regions like the NAc, BNST, and in particular the ARC, VMN and VTA that occur in sex- and energy status-dependent ways.
Collapse
Affiliation(s)
- Nikki Le
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Sarah Sayers
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Veronica Mata-Pacheco
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Edward J. Wagner
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| |
Collapse
|
8
|
Tu L, Fukuda M, Tong Q, Xu Y. The ventromedial hypothalamic nucleus: watchdog of whole-body glucose homeostasis. Cell Biosci 2022; 12:71. [PMID: 35619170 PMCID: PMC9134642 DOI: 10.1186/s13578-022-00799-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/25/2022] [Indexed: 02/06/2023] Open
Abstract
The brain, particularly the ventromedial hypothalamic nucleus (VMH), has been long known for its involvement in glucose sensing and whole-body glucose homeostasis. However, it is still not fully understood how the brain detects and responds to the changes in the circulating glucose levels, as well as brain-body coordinated control of glucose homeostasis. In this review, we address the growing evidence implicating the brain in glucose homeostasis, especially in the contexts of hypoglycemia and diabetes. In addition to neurons, we emphasize the potential roles played by non-neuronal cells, as well as extracellular matrix in the hypothalamus in whole-body glucose homeostasis. Further, we review the ionic mechanisms by which glucose-sensing neurons sense fluctuations of ambient glucose levels. We also introduce the significant implications of heterogeneous neurons in the VMH upon glucose sensing and whole-body glucose homeostasis, in which sex difference is also addressed. Meanwhile, research gaps have also been identified, which necessities further mechanistic studies in future.
Collapse
Affiliation(s)
- Longlong Tu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street #8066, Houston, TX, 77030, USA
| | - Makoto Fukuda
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street #8066, Houston, TX, 77030, USA
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street #8066, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
9
|
Pituitary adenylate cyclase-activating polypeptide type 1 receptor within the nucleus accumbens core mediates excessive alcohol drinking in alcohol-preferring rats. Neuropharmacology 2022; 212:109063. [PMID: 35460713 DOI: 10.1016/j.neuropharm.2022.109063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/19/2022] [Accepted: 04/12/2022] [Indexed: 12/15/2022]
Abstract
Alcohol use disorders (AUD) have a strong component of heritability; however, the neurobiological mechanisms mediating the propensity to consume excessive amounts of alcohol are still not well understood. Pituitary adenylate cyclase-activating polypeptide (PACAP), a highly conserved neuropeptide which exerts its effects mainly through the PAC1 receptor (PAC1R), has been suggested to be one of the mediators of the effects of drugs of abuse and alcohol. Here, we investigated the role of the PACAP/PAC1R system in excessive alcohol drinking in alcohol-preferring rats, an established animal model of AUD. Intracerebroventricular (i.c.v.) administration of the PAC1R antagonist PACAP(6-38) blocked excessive alcohol drinking and motivation to drink in Sardinian alcohol-preferring (Scr:sP) rats, without affecting water, saccharin, or sucrose intake. Notably, PACAP(6-38) did not affect ethanol responding in outbred Wistar rats. PACAP(6-38) also significantly reduced alcohol-seeking behavior under a second-order schedule of reinforcement. Using immunohistochemistry, a significant increase in the number of PAC1R positive cells was observed selectively in the nucleus accumbens (NAcc) Core of Scr:sP rats, compared to Wistar rats following alcohol drinking. Finally, excessive drinking in Scr:sP rats was suppressed by intra-NAcc Core, but not intra-NAcc Shell, PACAP(6-38), as well as by virally-mediated PAC1R knockdown in the NAcc Core. The present study shows that hyperactivity of the PACAP/PAC1R system specifically in the NAcc Core mediates excessive drinking of alcohol-preferring rats, and indicates that this system may represent a novel target for the treatment of AUD.
Collapse
|
10
|
Maunze B, Bruckner KW, Desai NN, Chen C, Chen F, Baker D, Choi S. Pituitary adenylate cyclase-activating polypeptide receptor activation in the hypothalamus recruits unique signaling pathways involved in energy homeostasis. Am J Physiol Endocrinol Metab 2022; 322:E199-E210. [PMID: 35001657 PMCID: PMC8897015 DOI: 10.1152/ajpendo.00320.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) exerts pleiotropic effects on ventromedial nuclei (VMN) of the hypothalamus and its control of feeding and energy expenditure through the type I PAC1 receptor (PAC1R). However, the endogenous role of PAC1Rs in the VMN and the downstream signaling responsible for PACAP's effects on energy balance are unknown. Numerous studies have revealed that PAC1Rs are coupled to both Gαs/adenylyl cyclase/protein kinase A (Gαs/AC/PKA) and Gαq/phospholipase C/protein kinase C (Gαq/PLC/PKC), while also undergoing trafficking following stimulation. To determine the endogenous role of PAC1Rs and downstream signaling that may explain PACAP's pleiotropic effects, we used RNA interference to knockdown VMN PAC1Rs and pharmacologically inhibited PKA, PKC, and PAC1R trafficking. Knocking down PAC1Rs increased meal sizes, reduced total number of meals, and induced body weight gain. Inhibition of either PKA or PKC alone in awake male Sprague-Dawley rats, attenuated PACAP's hypophagic and anorectic effects during the dark phase. However, PKA or PKC inhibition potentiated PACAP's thermogenic effects during the light phase. Analysis of locomotor activity revealed that PKA inhibition augmented PACAP's locomotor effects, whereas PKC inhibition had no effect. Finally, PACAP administration in the VMN induces surface PAC1R trafficking into the cytosol which was blocked by endocytosis inhibitors. Subsequently, inhibition of PAC1R trafficking into the cytosol attenuated PACAP-induced hypophagia. These results revealed that endogenous PAC1Rs uniquely engage PKA, PKC, and receptor trafficking to mediate PACAP's pleiotropic effects in VMN control of feeding and metabolism.NEW & NOTEWORTHY Endogenous PAC1 receptors, integral to VMN management of feeding behavior and body weight regulation, uniquely engage PKA, PKC, and receptor trafficking to mediate the hypothalamic ventromedial nuclei control of feeding and metabolism. PACAP appears to use different signaling mechanisms to regulate feeding behavior from its effects on metabolism.
Collapse
Affiliation(s)
- Brian Maunze
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin
| | | | - Nikhil Nilesh Desai
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin
| | - Christopher Chen
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin
| | - Fanghong Chen
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin
| | - David Baker
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin
| | - SuJean Choi
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin
| |
Collapse
|
11
|
Sureshkumar K, Saenz A, Ahmad SM, Lutfy K. The PACAP/PAC1 Receptor System and Feeding. Brain Sci 2021; 12:brainsci12010013. [PMID: 35053757 PMCID: PMC8773599 DOI: 10.3390/brainsci12010013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/13/2021] [Accepted: 12/20/2021] [Indexed: 12/18/2022] Open
Abstract
Pituitary adenylyl cyclase activating polypeptide (PACAP) belongs to the vasoactive intestinal polypeptide (VIP)/secretin/glucagon superfamily. PACAP is present in two forms (PACAP-38 and PACAP-27) and binds to three guanine-regulatory (G) protein-coupled receptors (PAC1, VPAC1, and VPAC2). PACAP is expressed in the central and peripheral nervous systems, with high PACAP levels found in the hypothalamus, a brain region involved in feeding and energy homeostasis. PAC1 receptors are high-affinity and PACAP-selective receptors, while VPAC1 and VPAC2 receptors show a comparable affinity to PACAP and VIP. PACAP and its receptors are expressed in the central and peripheral nervous systems with moderate to high expression in the hypothalamus, amygdala, and other limbic structures. Consistent with their expression, PACAP is involved in several physiological responses and pathological states. A growing body of literature suggests that PACAP regulates food intake in laboratory animals. However, there is no comprehensive review of the literature on this topic. Thus, the purpose of this article is to review the literature regarding the role of PACAP and its receptors in food intake regulation and to synthesize how PACAP exerts its anorexic effects in different brain regions. To achieve this goal, we searched PubMed and reviewed 68 articles regarding the regulatory action of PACAP on food intake. Here, we present the literature regarding the effect of exogenous PACAP on feeding and the role of endogenous PACAP in this process. We also provide evidence regarding the effect of PACAP on the homeostatic and hedonic aspects of food intake, the neuroanatomical sites where PACAP exerts its regulatory action, which PACAP receptors may be involved, and the role of various signaling pathways and neurotransmitters in hypophagic effects of PACAP.
Collapse
Affiliation(s)
- Keerthana Sureshkumar
- UCLA College of Letters and Sciences, University of California, 612 Charles E Young Dr. South, Los Angeles, CA 90095, USA;
| | - Andrea Saenz
- College of Pharmacy, Western University of Health Sciences, 309 East Second Street, Pomona, CA 91766, USA; (A.S.); (S.M.A.)
| | - Syed M. Ahmad
- College of Pharmacy, Western University of Health Sciences, 309 East Second Street, Pomona, CA 91766, USA; (A.S.); (S.M.A.)
| | - Kabirullah Lutfy
- College of Pharmacy, Western University of Health Sciences, 309 East Second Street, Pomona, CA 91766, USA; (A.S.); (S.M.A.)
- Correspondence: ; Tel.: +1-(909)-469-5481
| |
Collapse
|
12
|
Minnig MA, Park T, Echeveste Sanchez M, Cottone P, Sabino V. Viral-Mediated Knockdown of Nucleus Accumbens Shell PAC1 Receptor Promotes Excessive Alcohol Drinking in Alcohol-Preferring Rats. Front Behav Neurosci 2021; 15:787362. [PMID: 34924973 PMCID: PMC8678417 DOI: 10.3389/fnbeh.2021.787362] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/08/2021] [Indexed: 01/04/2023] Open
Abstract
Alcohol use disorder (AUD) is a chronic, relapsing disorder whose genetic and environmental susceptibility components are not fully understood. Neuropeptidergic signaling has been repeatedly implicated in modulating excessive alcohol drinking, especially within sub-regions of the striatum. Here, we investigated the potential involvement of the selective receptor for pituitary adenylate cyclase-activating polypeptide (PACAP), PAC1R, in the nucleus accumbens shell (NAcc Shell) in excessive alcohol drinking in alcohol-preferring rats, an established animal model of the genetic propensity for alcoholism. Scr:sP alcohol-preferring rats were trained to operantly self-administer alcohol and then either an AAV virus short-hairpin RNA (shRNA) targeted to knockdown PAC1R, or an AAV control virus were microinfused into the NAcc Shell. NAcc Shell PAC1R shRNA knockdown virus was confirmed to significantly decrease PAC1R levels in the NAcc Shell. The effects of NAcc Shell PAC1R shRNA knockdown on ethanol self-administration were investigated using a Fixed Ratio (FR) 1 and a Progressive Ratio (PR) schedule of reinforcement. The effect of PAC1R knockdown on self-administration of an alternative reinforcer, saccharin, was also assessed. The results showed that the reduction in PAC1R in the NAcc Shell led to excessive ethanol drinking, increased preference for ethanol, and higher motivation to drink. NAcc Shell PAC1R shRNA knockdown did not comparably increase saccharin self-administration, suggesting selectivity of action. These data suggest that NAcc Shell PAC1R may serves as a "brake" on alcohol drinking, and thereby the loss of function of PAC1R leads to excessive alcohol consumption. Therefore, the PACAP/PAC1R system may represent a novel target for the treatment of AUD.
Collapse
Affiliation(s)
| | | | | | | | - Valentina Sabino
- Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
13
|
Le N, Hernandez J, Gastelum C, Perez L, Vahrson I, Sayers S, Wagner EJ. Pituitary Adenylate Cyclase Activating Polypeptide Inhibits A 10 Dopamine Neurons and Suppresses the Binge-like Consumption of Palatable Food. Neuroscience 2021; 478:49-64. [PMID: 34597709 DOI: 10.1016/j.neuroscience.2021.09.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 10/20/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) binds to PACAP-specific (PAC1) receptors in multiple hypothalamic areas, especially those regulating energy balance. PACAP neurons in the ventromedial nucleus (VMN) exert anorexigenic effects within the homeostatic energy balance circuitry. Since PACAP can also reduce the consumption of palatable food, we tested the hypothesis that VMN PACAP neurons project to the ventral tegmental area (VTA) to inhibit A10 dopamine neurons via PAC1 receptors and KATP channels, and thereby suppress binge-like consumption. We performed electrophysiological recordings in mesencephalic slices from male PACAP-Cre and tyrosine hydroxylase (TH)-Cre mice. Initially, we injected PACAP (30 pmol) into the VTA, where it suppressed binge intake in wildtype male but not female mice. Subsequent tract tracing studies uncovered projections of VMN PACAP neurons to the VTA. Optogenetic stimulation of VMN PACAP neurons in voltage clamp induced an outward current and increase in conductance in VTA neurons, and a hyperpolarization and decrease in firing in current clamp. These effects were markedly attenuated by the KATP channel blocker tolbutamide (100 μM) and PAC1 receptor antagonist PACAP6-38 (200 nM). In recordings from A10 dopamine neurons in TH-Cre mice, we replicated the outward current by perfusing PACAP1-38 (100 nM). This response was again completely blocked by tolbutamide and PACAP6-38, and associated with a hyperpolarization and decrease in firing. These findings demonstrate that PACAP activates PAC1 receptors and KATP channels to inhibit A10 dopamine neurons and sex-dependently suppress binge-like consumption. Accordingly, they advance our understanding of how PACAP regulates energy homeostasis via the hedonic energy balance circuitry.
Collapse
Affiliation(s)
- Nikki Le
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Jennifer Hernandez
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Cassandra Gastelum
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Lynnea Perez
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Isabella Vahrson
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Sarah Sayers
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Edward J Wagner
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA; College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA.
| |
Collapse
|
14
|
Martins-Oliveira M, Tavares I, Goadsby PJ. Was it something I ate? Understanding the bidirectional interaction of migraine and appetite neural circuits. Brain Res 2021; 1770:147629. [PMID: 34428465 DOI: 10.1016/j.brainres.2021.147629] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 12/18/2022]
Abstract
Migraine attacks can involve changes of appetite: while fasting or skipping meals are often reported triggers in susceptible individuals, hunger or food craving are reported in the premonitory phase. Over the last decade, there has been a growing interest and recognition of the importance of studying these overlapping fields of neuroscience, which has led to novel findings. The data suggest additional studies are needed to unravel key neurobiological mechanisms underlying the bidirectional interaction between migraine and appetite. Herein, we review information about the metabolic migraine phenotype and explore migraine therapeutic targets that have a strong input on appetite neuronal circuits, including the calcitonin gene-related peptide (CGRP), the pituitary adenylate cyclase-activating polypeptide (PACAP) and the orexins. Furthermore, we focus on potential therapeutic peptide targets that are involved in regulation of feeding and play a role in migraine pathophysiology, such as neuropeptide Y, insulin, glucagon and leptin. We then examine the orexigenic - anorexigenic circuit feedback loop and explore glucose metabolism disturbances. Additionally, it is proposed a different perspective on the most reported feeding-related trigger - skipping meals - as well as a link between contrasting feeding behaviors (skipping meals vs food craving). Our review aims to increase awareness of migraine through the lens of appetite neurobiology in order to improve our understanding of the earlier phase of migraine, encourage better studies and cross-disciplinary collaborations, and provide novel migraine-specific therapeutic opportunities.
Collapse
Affiliation(s)
- Margarida Martins-Oliveira
- Headache Group, Wolfson Centre for Age-Related Disease, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK; Nutrition and Metabolism Department, NOVA Medical School, Faculdade de Ciências Médicas de Lisboa, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisbon, Portugal.
| | - Isaura Tavares
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; Institute of Investigation and Innovation in Health (i3S), University of Porto, Portugal.
| | - Peter J Goadsby
- Headache Group, Wolfson Centre for Age-Related Disease, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK; Department of Neurology, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
15
|
Gastelum C, Perez L, Hernandez J, Le N, Vahrson I, Sayers S, Wagner EJ. Adaptive Changes in the Central Control of Energy Homeostasis Occur in Response to Variations in Energy Status. Int J Mol Sci 2021; 22:2728. [PMID: 33800452 PMCID: PMC7962960 DOI: 10.3390/ijms22052728] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/15/2022] Open
Abstract
Energy homeostasis is regulated in coordinate fashion by the brain-gut axis, the homeostatic energy balance circuitry in the hypothalamus and the hedonic energy balance circuitry comprising the mesolimbcortical A10 dopamine pathway. Collectively, these systems convey and integrate information regarding nutrient status and the rewarding properties of ingested food, and formulate it into a behavioral response that attempts to balance fluctuations in consumption and food-seeking behavior. In this review we start with a functional overview of the homeostatic and hedonic energy balance circuitries; identifying the salient neural, hormonal and humoral components involved. We then delve into how the function of these circuits differs in males and females. Finally, we turn our attention to the ever-emerging roles of nociceptin/orphanin FQ (N/OFQ) and pituitary adenylate cyclase-activating polypeptide (PACAP)-two neuropeptides that have garnered increased recognition for their regulatory impact in energy homeostasis-to further probe how the imposed regulation of energy balance circuitry by these peptides is affected by sex and altered under positive (e.g., obesity) and negative (e.g., fasting) energy balance states. It is hoped that this work will impart a newfound appreciation for the intricate regulatory processes that govern energy homeostasis, as well as how recent insights into the N/OFQ and PACAP systems can be leveraged in the treatment of conditions ranging from obesity to anorexia.
Collapse
Affiliation(s)
- Cassandra Gastelum
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (C.G.); (L.P.); (J.H.); (N.L.); (I.V.); (S.S.)
| | - Lynnea Perez
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (C.G.); (L.P.); (J.H.); (N.L.); (I.V.); (S.S.)
| | - Jennifer Hernandez
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (C.G.); (L.P.); (J.H.); (N.L.); (I.V.); (S.S.)
| | - Nikki Le
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (C.G.); (L.P.); (J.H.); (N.L.); (I.V.); (S.S.)
| | - Isabella Vahrson
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (C.G.); (L.P.); (J.H.); (N.L.); (I.V.); (S.S.)
| | - Sarah Sayers
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (C.G.); (L.P.); (J.H.); (N.L.); (I.V.); (S.S.)
| | - Edward J. Wagner
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (C.G.); (L.P.); (J.H.); (N.L.); (I.V.); (S.S.)
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
16
|
Roth E, Benoit S, Quentin B, Lam B, Will S, Ma M, Heeley N, Darwish T, Shrestha Y, Gribble F, Reimann F, Pshenichnaya I, Yeo G, Baker DJ, Trevaskis JL, Blouet C. Behavioural and neurochemical mechanisms underpinning the feeding-suppressive effect of GLP-1/CCK combinatorial therapy. Mol Metab 2021; 43:101118. [PMID: 33221554 PMCID: PMC7720077 DOI: 10.1016/j.molmet.2020.101118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVES Combinatorial therapies are under intense investigation to develop more efficient anti-obesity drugs; however, little is known about how they act in the brain to produce enhanced anorexia and weight loss. The goal of this study was to identify the brain sites and neuronal populations engaged during the co-administration of GLP-1R and CCK1R agonists, an efficient combination therapy in obese rodents. METHODS We measured acute and long-term feeding and body weight responses and neuronal activation patterns throughout the neuraxis and in specific neuronal subsets in response to GLP-1R and CCK1R agonists administered alone or in combination in lean and high-fat diet fed mice. We used PhosphoTRAP to obtain unbiased molecular markers for neuronal populations selectively activated by the combination of the two agonists. RESULTS The initial anorectic response to GLP-1R and CCK1R co-agonism was mediated by a reduction in meal size, but over a few hours, a reduction in meal number accounted for the sustained feeding suppressive effects. The nucleus of the solitary tract (NTS) is one of the few brain sites where GLP-1R and CCK1R signalling interact to produce enhanced neuronal activation. None of the previously categorised NTS neuronal subpopulations relevant to feeding behaviour were implicated in this increased activation. However, we identified NTS/AP Calcrl+ neurons as treatment targets. CONCLUSIONS Collectively, these studies indicated that circuit-level integration of GLP-1R and CCK1R co-agonism in discrete brain nuclei including the NTS produces enhanced rapid and sustained appetite suppression and weight loss.
Collapse
Affiliation(s)
- Emma Roth
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 OQQ, UK
| | - Simon Benoit
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 OQQ, UK
| | - Baptiste Quentin
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 OQQ, UK
| | - Brian Lam
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 OQQ, UK
| | - Sarah Will
- Cardiovascular, Renal, and Metabolic Diseases, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Marcella Ma
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 OQQ, UK
| | - Nick Heeley
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 OQQ, UK
| | - Tamana Darwish
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 OQQ, UK
| | - Yashaswi Shrestha
- Early Oncology, Translational Medicine, MedImmune LLC, Gaithersburg, MD 20878, USA
| | - Fiona Gribble
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 OQQ, UK
| | - Frank Reimann
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 OQQ, UK
| | | | - Giles Yeo
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 OQQ, UK
| | - David J Baker
- Cardiovascular, Renal and Metabolic Diseases, MedImmune Ltd., Cambridge, UK
| | - James L Trevaskis
- Cardiovascular, Renal and Metabolic Diseases, MedImmune Ltd., Cambridge, UK
| | - Clemence Blouet
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 OQQ, UK.
| |
Collapse
|
17
|
Chang R, Hernandez J, Gastelum C, Guadagno K, Perez L, Wagner EJ. Pituitary Adenylate Cyclase-Activating Polypeptide Excites Proopiomelanocortin Neurons: Implications for the Regulation of Energy Homeostasis. Neuroendocrinology 2021; 111:45-69. [PMID: 32028278 DOI: 10.1159/000506367] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/30/2020] [Indexed: 11/19/2022]
Abstract
OBJECTIVE We examined whether pituitary adenylate cyclase-activating polypeptide (PACAP) excites proopiomelanocortin (POMC) neurons via PAC1 receptor mediation and transient receptor potential cation (TRPC) channel activation. METHODS Electrophysiological recordings were done in slices from both intact male and ovariectomized (OVX) female PACAP-Cre mice and eGFP-POMC mice. RESULTS In recordings from POMC neurons in eGFP-POMC mice, PACAP induced a robust inward current and increase in conductance in voltage clamp, and a depolarization and increase in firing in current clamp. These postsynaptic actions were abolished by inhibitors of the PAC1 receptor, TRPC channels, phospholipase C, phosphatidylinositol-3-kinase, and protein kinase C. Estradiol augmented the PACAP-induced inward current, depolarization, and increased firing, which was abrogated by estrogen receptor (ER) antagonists. In optogenetic recordings from POMC neurons in PACAP-Cre mice, high-frequency photostimulation induced inward currents, depolarizations, and increased firing that were significantly enhanced by Gq-coupled membrane ER signaling in an ER antagonist-sensitive manner. Importantly, the PACAP-induced excitation of POMC neurons was notably reduced in obese, high-fat (HFD)-fed males. In vivo experiments revealed that intra-arcuate nucleus (ARC) PACAP as well as chemogenetic and optogenetic stimulation of ventromedial nucleus (VMN) PACAP neurons produced a significant decrease in energy intake accompanied by an increase in energy expenditure, effects blunted by HFD in males and partially potentiated by estradiol in OVX females. CONCLUSIONS These findings reveal that the PACAP-induced activation of PAC1 receptor and TRPC5 channels at VMN PACAP/ARC POMC synapses is potentiated by estradiol and attenuated under conditions of diet-induced obesity/insulin resistance. As such, they advance our understanding of how PACAP regulates the homeostatic energy balance circuitry under normal and pathophysiological circumstances.
Collapse
Affiliation(s)
- Rachel Chang
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, California, USA
| | - Jennifer Hernandez
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Cassandra Gastelum
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, California, USA
| | - Kaitlyn Guadagno
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Lynnea Perez
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, California, USA
| | - Edward J Wagner
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, California, USA,
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA,
| |
Collapse
|
18
|
McMillan TR, Forster MAM, Short LI, Rudecki AP, Cline DL, Gray SL. Melanotan II, a melanocortin agonist, partially rescues the impaired thermogenic capacity of pituitary adenylate cyclase-activating polypeptide deficient mice. Exp Physiol 2020; 106:427-437. [PMID: 33332767 DOI: 10.1113/ep088838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 11/27/2020] [Indexed: 12/16/2022]
Abstract
NEW FINDINGS What is the central question of this study? Can chronic treatment of pituitary adenylate cyclase-activating polypeptide (PACAP) deficient mice with the melanocortin agonist melanotan II during cold acclimation rescue the impaired thermogenic capacity previously observed in PACAP deficient mice? What is the main finding and its importance? Using a genetic model of PACAP deficiency, this study provides evidence that PACAP acts upstream of the melanocortin system in regulating sympathetic nerve activity to brown adipose tissue in mice. ABSTRACT Impaired adipose tissue function in obesity, including reduced thermogenic potential, has detrimental consequences for metabolic health. Hormonal regulation of adaptive thermogenesis is being explored as a potential therapeutic target for human obesity. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide expressed in nuclei of the hypothalamus known to regulate energy expenditure, and functional studies reveal a role for PACAP in the central regulation of thermogenesis, although mechanisms are not well understood. We hypothesized that PACAP acts upstream of the melanocortin system to regulate sympathetic nerve activity to stimulate thermogenesis. To assess this, female PACAP-/- and PACAP+/+ mice were given daily peripheral injections of a melanocortin receptor agonist, melanotan II (MTII), for 3 weeks during cold acclimation, and the effect of MTII on thermogenic capacity and adipose tissue remodelling was examined by physiological and histological analyses. MTII partially rescued the impaired thermogenic capacity in PACAP-/- mice as compared to PACAP+/+ mice as determined by measuring noradrenaline-induced metabolic rate. In addition, MTII treatment during cold acclimation corrected the previously identified deficit in lipid utilization in response to adrenergic stimulation in PACAP-/- null mice, suggesting impaired lipid mobilization may contribute to the impaired thermogenic capacity of PACAP-/- mice. Results presented here provide physiological evidence to suggest that PACAP acts upstream of melanocortin receptors to facilitate sympathetically induced mechanisms of adaptive thermogenesis in response to cold acclimation.
Collapse
Affiliation(s)
- Thecla Rae McMillan
- Northern Medical Program, University of Northern British Columbia, Prince George, British Columbia, Canada
| | - Maeghan A M Forster
- Northern Medical Program, University of Northern British Columbia, Prince George, British Columbia, Canada
| | - Landon I Short
- Northern Medical Program, University of Northern British Columbia, Prince George, British Columbia, Canada
| | - Alexander P Rudecki
- Northern Medical Program, University of Northern British Columbia, Prince George, British Columbia, Canada
| | - Daemon L Cline
- Northern Medical Program, University of Northern British Columbia, Prince George, British Columbia, Canada
| | - Sarah L Gray
- Northern Medical Program, University of Northern British Columbia, Prince George, British Columbia, Canada
| |
Collapse
|
19
|
Fang Y, Ren R, Shi H, Huang L, Lenahan C, Lu Q, Tang L, Huang Y, Tang J, Zhang J, Zhang JH. Pituitary Adenylate Cyclase-Activating Polypeptide: A Promising Neuroprotective Peptide in Stroke. Aging Dis 2020; 11:1496-1512. [PMID: 33269103 PMCID: PMC7673855 DOI: 10.14336/ad.2020.0626] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 06/26/2020] [Indexed: 12/11/2022] Open
Abstract
The search for viable, effective treatments for acute stroke continues to be a global priority due to the high mortality and morbidity. Current therapeutic treatments have limited effects, making the search for new treatments imperative. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a well-established cytoprotective neuropeptide that participates in diverse neural physiological and pathological activities, such as neuronal proliferation, differentiation, and migration, as well as neuroprotection. It is considered a promising treatment in numerous neurological diseases. Thus, PACAP bears potential as a new therapeutic strategy for stroke treatment. Herein, we provide an overview pertaining to the current knowledge of PACAP, its receptors, and its potential neuroprotective role in the setting of stroke, as well as various mechanisms of neuroprotection involving ionic homeostasis, excitotoxicity, cell edema, oxidative stress, inflammation, and cell death, as well as the route of PACAP administration.
Collapse
Affiliation(s)
- Yuanjian Fang
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Reng Ren
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hui Shi
- 2Department of Neurosurgery, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Lei Huang
- 3Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA.,4Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Cameron Lenahan
- 3Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA.,4Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA.,5Burrell College of Osteopathic Medicine, Las Cruces, NM, USA
| | - Qin Lu
- 6Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China
| | - Lihui Tang
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yi Huang
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiping Tang
- 3Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA.,4Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA.,7Department of Anesthesiology, Loma Linda University, Loma Linda, CA, USA
| | - Jianmin Zhang
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - John H Zhang
- 3Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA.,4Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA.,7Department of Anesthesiology, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
20
|
Circulating PACAP peptide and PAC1R genotype as possible transdiagnostic biomarkers for anxiety disorders in women: a preliminary study. Neuropsychopharmacology 2020; 45:1125-1133. [PMID: 31910434 PMCID: PMC7235237 DOI: 10.1038/s41386-020-0604-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 11/30/2019] [Accepted: 12/30/2019] [Indexed: 01/04/2023]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP, gene Adcyap1) is a neuropeptide and hormone thought to play a critical role in stress response (Stroth et al., Ann NY Acad Sci 1220:49-59, 2011; Hashimoto et al., Curr Pharm Des 17:985-989, 2011). Research in humans implicates PACAP as a useful biomarker for the severity of psychiatric symptoms in response to psychological stressors, and work in rodent models suggests that PACAP manipulation exerts downstream effects on peripheral hormones and behaviors linked to the stress response, providing a potential therapeutic target. Prior work has also suggested a potential sex difference in PACAP effects due to differential estrogen regulation of this pathway. Therefore, we examined serum PACAP and associated PAC1R genotype in a cohort of males and females with a primary diagnosis of generalized anxiety disorder (GAD) and nonpsychiatric controls. We found that, while circulating hormone levels were not associated with a GAD diagnosis overall (p = 0.19, g = 0.25), PACAP may be associated with GAD in females (p = 0.04, g = 0.33). Additionally, among patients with GAD, the risk genotype identified in the PTSD literature (rs2267735, CC genotype) was associated with higher somatic anxiety symptom severity in females but lower somatic anxiety symptom severity in males (-3.27, 95%CI [-5.76, -0.77], adjusted p = 0.03). Taken together, the associations between the risk genotype, circulating PACAP, and somatic anxiety severity were stronger among females than males. These results indicate a potential underlying biological etiology for sex differences in stress-related anxiety disorders that warrants further study.
Collapse
|
21
|
Ceddia RP, Collins S. A compendium of G-protein-coupled receptors and cyclic nucleotide regulation of adipose tissue metabolism and energy expenditure. Clin Sci (Lond) 2020; 134:473-512. [PMID: 32149342 PMCID: PMC9137350 DOI: 10.1042/cs20190579] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/17/2020] [Accepted: 02/24/2020] [Indexed: 12/15/2022]
Abstract
With the ever-increasing burden of obesity and Type 2 diabetes, it is generally acknowledged that there remains a need for developing new therapeutics. One potential mechanism to combat obesity is to raise energy expenditure via increasing the amount of uncoupled respiration from the mitochondria-rich brown and beige adipocytes. With the recent appreciation of thermogenic adipocytes in humans, much effort is being made to elucidate the signaling pathways that regulate the browning of adipose tissue. In this review, we focus on the ligand-receptor signaling pathways that influence the cyclic nucleotides, cAMP and cGMP, in adipocytes. We chose to focus on G-protein-coupled receptor (GPCR), guanylyl cyclase and phosphodiesterase regulation of adipocytes because they are the targets of a large proportion of all currently available therapeutics. Furthermore, there is a large overlap in their signaling pathways, as signaling events that raise cAMP or cGMP generally increase adipocyte lipolysis and cause changes that are commonly referred to as browning: increasing mitochondrial biogenesis, uncoupling protein 1 (UCP1) expression and respiration.
Collapse
Affiliation(s)
- Ryan P Ceddia
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, U.S.A
| | - Sheila Collins
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, U.S.A
| |
Collapse
|
22
|
Köves K, Szabó E, Kántor O, Heinzlmann A, Szabó F, Csáki Á. Current State of Understanding of the Role of PACAP in the Hypothalamo-Hypophyseal Gonadotropin Functions of Mammals. Front Endocrinol (Lausanne) 2020; 11:88. [PMID: 32210912 PMCID: PMC7067695 DOI: 10.3389/fendo.2020.00088] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 02/11/2020] [Indexed: 01/25/2023] Open
Abstract
PACAP was discovered 30 years ago in Dr. Akira Arimura's laboratory. In the past three decades since then, it has become evident that this peptide plays numerous crucial roles in mammalian organisms. The most important functions of PACAP are the following: 1. neurotransmitter, 2. neuromodulator, 3. hypophysiotropic hormone, 4. neuroprotector. This paper reviews the accumulated data regarding the distribution of PACAP and its receptors in the mammalian hypothalamus and pituitary gland, the role of PACAP in the gonadotropin hormone secretion of females and males. The review also summarizes the interaction between PACAP, GnRH, and sex steroids as well as hypothalamic peptides including kisspeptin. The possible role of PACAP in reproductive functions through the biological clock is also discussed. Finally, the significance of PACAP in the hypothalamo-hypophysial system is considered and the facts missing, that would help better understand the function of PACAP in this system, are also highlighted.
Collapse
Affiliation(s)
- Katalin Köves
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Enikő Szabó
- Department of Conservative Dentistry, Faculty of Dentistry, Semmelweis University, Budapest, Hungary
| | - Orsolya Kántor
- Department of Molecular Embryology, Medical Faculty, Institute of Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany
| | - Andrea Heinzlmann
- Department of Anatomy and Histology, University of Veterinary Sciences, Budapest, Hungary
| | - Flóra Szabó
- Department of Pediatrics, Virginia Commonwealth University, Richmond, VA, United States
| | - Ágnes Csáki
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
23
|
Pituitary Adenylate Cyclase-Activating Polypeptide in the Ventromedial Hypothalamus Is Responsible for Food Intake Behavior by Modulating the Expression of Agouti-Related Peptide in Mice. Mol Neurobiol 2020; 57:2101-2114. [DOI: 10.1007/s12035-019-01864-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 12/22/2019] [Indexed: 12/18/2022]
|
24
|
Pleiotropic pituitary adenylate cyclase-activating polypeptide (PACAP): Novel insights into the role of PACAP in eating and drug intake. Brain Res 2019; 1729:146626. [PMID: 31883848 PMCID: PMC6953419 DOI: 10.1016/j.brainres.2019.146626] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 12/20/2019] [Accepted: 12/24/2019] [Indexed: 01/30/2023]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) was discovered thirty years ago, but its role in eating and drug use disorders has only recently begun to be investigated. The present review develops the hypothesis that, although PACAP normally functions to tightly regulate intake, inhibiting it through negative feedback, this relationship can become dysregulated with the development of dependence, such that PACAP instead acts through positive feedback to promote excessive intake. We propose that repeated exposure to palatable food and drugs of abuse can alter the downstream responses of specific populations of neurons to stimulation by PACAP, leading to the perpetuation of the addiction cycle. Thus, this review will first describe published literature on homeostatic food intake, which shows that PACAP suppresses food intake, while its levels are themselves increased by overfeeding. Next, it will present literature on palatable food, cocaine, alcohol, and nicotine, which overall demonstrates that PACAP in specific limbic brain regions can promote their seeking and intake and itself is stimulated by their intake. Then, it will present literature on affective behavior, which shows that chronic stress increases levels of PACAP, which then promotes anxiety and depression, factors that can trigger substance seeking. Finally, the review will address mechanisms through which chronic substance exposure may dysregulate the PACAP system, proposing that it alters expression of PACAP receptor splice variants. While many questions remain to be addressed, the current evidence suggests that PACAP could be a viable medication target for the treatment of binge eating and drug and alcohol use disorders.
Collapse
|
25
|
Liao C, de Molliens MP, Schneebeli ST, Brewer M, Song G, Chatenet D, Braas KM, May V, Li J. Targeting the PAC1 Receptor for Neurological and Metabolic Disorders. Curr Top Med Chem 2019; 19:1399-1417. [PMID: 31284862 PMCID: PMC6761004 DOI: 10.2174/1568026619666190709092647] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 12/23/2018] [Accepted: 12/26/2018] [Indexed: 12/16/2022]
Abstract
The pituitary adenylate cyclase-activating polypeptide (PACAP)-selective PAC1 receptor (PAC1R, ADCYAP1R1) is a member of the vasoactive intestinal peptide (VIP)/secretin/glucagon family of G protein-coupled receptors (GPCRs). PAC1R has been shown to play crucial roles in the central and peripheral nervous systems. The activation of PAC1R initiates diverse downstream signal transduction pathways, including adenylyl cyclase, phospholipase C, MEK/ERK, and Akt pathways that regulate a number of physiological systems to maintain functional homeostasis. Accordingly, at times of tissue injury or insult, PACAP/PAC1R activation of these pathways can be trophic to blunt or delay apoptotic events and enhance cell survival. Enhancing PAC1R signaling under these conditions has the potential to mitigate cellular damages associated with cerebrovascular trauma (including stroke), neurodegeneration (such as Parkinson's and Alzheimer's disease), or peripheral organ insults. Conversely, maladaptive PACAP/PAC1R signaling has been implicated in a number of disorders, including stressrelated psychopathologies (i.e., depression, posttraumatic stress disorder, and related abnormalities), chronic pain and migraine, and metabolic diseases; abrogating PAC1R signaling under these pathological conditions represent opportunities for therapeutic intervention. Given the diverse PAC1R-mediated biological activities, the receptor has emerged as a relevant pharmaceutical target. In this review, we first describe the current knowledge regarding the molecular structure, dynamics, and function of PAC1R. Then, we discuss the roles of PACAP and PAC1R in the activation of a variety of signaling cascades related to the physiology and diseases of the nervous system. Lastly, we examine current drug design and development of peptides and small molecules targeting PAC1R based on a number of structure- activity relationship studies and key pharmacophore elements. At present, the rational design of PAC1R-selective peptide or small-molecule therapeutics is largely hindered by the lack of structural information regarding PAC1R activation mechanisms, the PACAP-PAC1R interface, and the core segments involved in receptor activation. Understanding the molecular basis governing the PACAP interactions with its different cognate receptors will undoubtedly provide a basis for the development and/or refinement of receptor-selective therapeutics.
Collapse
Affiliation(s)
- Chenyi Liao
- Department of Chemistry, University of Vermont, Burlington, VT 05405, United States
| | | | - Severin T Schneebeli
- Department of Chemistry, University of Vermont, Burlington, VT 05405, United States
| | - Matthias Brewer
- Department of Chemistry, University of Vermont, Burlington, VT 05405, United States
| | - Gaojie Song
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - David Chatenet
- INRS - Institut Armand-Frappier, 531 boul. des Prairies, Laval, QC H7V 1B7, Canada
| | - Karen M Braas
- Department of Neurological Sciences, University of Vermont, Larner College of Medicine, 149 Beaumont Avenue, Burlington, VT 05405, United States
| | - Victor May
- Department of Neurological Sciences, University of Vermont, Larner College of Medicine, 149 Beaumont Avenue, Burlington, VT 05405, United States
| | - Jianing Li
- Department of Chemistry, University of Vermont, Burlington, VT 05405, United States
| |
Collapse
|
26
|
Hurley MM, Anderson EM, Chen C, Maunze B, Hess EM, Block ME, Patel N, Cooper Z, McCoy R, Dabra T, Conley W, Reilly MJ, Hearing M, Choi S. Acute Blockade of PACAP-Dependent Activity in the Ventromedial Nucleus of the Hypothalamus Disrupts Leptin-Induced Behavioral and Molecular Changes in Rats. Neuroendocrinology 2019; 110:271-281. [PMID: 31167202 PMCID: PMC6895395 DOI: 10.1159/000501337] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 06/05/2019] [Indexed: 12/18/2022]
Abstract
Leptin signaling pathways, stemming primarily from the hypothalamus, are necessary for maintaining normal energy homeostasis and body weight. In both rodents and humans, dysregulation of leptin signaling leads to morbid obesity and diabetes. Since leptin resistance is considered a primary factor underlying obesity, understanding the regulation of leptin signaling could lead to therapeutic tools and provide insights into the causality of obesity. While leptin actions in some hypothalamic regions such as the arcuate nuclei have been characterized, less is known about leptin activity in the hypothalamic ventromedial nuclei (VMN). Recently, pituitary adenylate cyclase-activating polypeptide (PACAP) has been shown to reduce feeding behavior and alter metabolism when administered into the VMN in a pattern similar to that of leptin. In the current study, we examined whether leptin and PACAP actions in the VMN share overlapping pathways in the regulation of energy balance. Interestingly, PACAP administration into the VMN increased STAT3 phosphorylation and SOCS3 mRNA expression, both of which are hallmarks of leptin receptor activation. In addition, BDNF mRNA expression in the VMN was increased by both leptin and PACAP administration. Moreover, antagonizing PACAP receptors fully reversed the behavioral and cellular effects of leptin injections into the VMN. Electrophysiological studies further illustrated that leptin-induced effects on VMN neurons were blocked by antagonizing PACAP receptors. We conclude that leptin dependency on PACAP signaling in the VMN suggests a potential common signaling cascade, allowing a tonically and systemically secreted neuropeptide to be more precisely regulated by central neuropeptides.
Collapse
Affiliation(s)
- Matthew M Hurley
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Eden M Anderson
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Christopher Chen
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Brian Maunze
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Evan M Hess
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Megan E Block
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Neerali Patel
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Zane Cooper
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Riley McCoy
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Tanya Dabra
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - William Conley
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Michael J Reilly
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Matthew Hearing
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - SuJean Choi
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA,
| |
Collapse
|
27
|
Nakamachi T, Tanigawa A, Konno N, Shioda S, Matsuda K. Expression Patterns of PACAP and PAC1R Genes and Anorexigenic Action of PACAP1 and PACAP2 in Zebrafish. Front Endocrinol (Lausanne) 2019; 10:227. [PMID: 31031705 PMCID: PMC6473066 DOI: 10.3389/fendo.2019.00227] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 03/21/2019] [Indexed: 11/13/2022] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide with potent suppressive effects on feeding behavior in rodents, chicken, and goldfish. Teleost fish express two PACAPs (PACAP1, encoded by the adcyap1a gene, and PACAP2, encoded by the adcyap1b gene) and two PACAP receptors (PAC1Rs; PAC1Ra, encoded by the adcyap1r1a gene, and PAC1Rb, encoded by the adcyap1r1b gene). However, the mRNA expression patterns of the two PACAPs and PAC1Rs, and the influence and relationship of the two PACAPs on feeding behavior in teleost fish remains unclear. Therefore, we first examined mRNA expression patterns of PACAP and PAC1R in tissue and brain. All PACAP and PAC1Rs mRNAs were dominantly expressed in the zebrafish brain. However, adcyap1a mRNA was also detected in the gut and testis. In the brain, adcyap1b and adcyap1r1a mRNA levels were greater than that of adcyap1a and adcyap1r1b, respectively. Moreover, adcyap1b and adcyap1r1a mRNA were dominantly expressed in telencephalon and diencephalon. The highest adcyap1a mRNA levels were detected in the brain stem and diencephalon, while the highest levels of adcyap1r1b were detected in the cerebellum. To clarify the relationship between PACAP and feeding behavior in the zebrafish, the effects of zebrafish (zf) PACAP1 or zfPACAP2 intracerebroventricular (ICV) injection were examined on food intake, and changes in PACAP mRNA levels were assessed against feeding status. Food intake was significantly decreased by ICV injection of zfPACAP1 (2 pmol/g body weight), zfPACAP2 (2 or 20 pmol/g body weight), or mammalian PACAP (2 or 20 pmol/g). Meanwhile, the PACAP injection group did not change locomotor activity. Real-time PCR showed adcyap1 mRNA levels were significantly increased at 2 and 3 h after feeding compared with the pre-feeding level, but adcyap1b, adcyap1r1a, and adcyap1r1b mRNA levels did not change after feeding. These results suggest that the expression levels and distribution of duplicated PACAP and PAC1R genes are different in zebrafish, but the anorexigenic effects of PACAP are similar to those seen in other vertebrates.
Collapse
Affiliation(s)
- Tomoya Nakamachi
- Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan
- *Correspondence: Tomoya Nakamachi
| | - Ayano Tanigawa
- Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan
| | - Norifumi Konno
- Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan
| | - Seiji Shioda
- Innovative Drug Discovery, Global Research Center for Innovative Life Science, Hoshi University, Tokyo, Japan
| | - Kouhei Matsuda
- Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan
| |
Collapse
|
28
|
Adipose Tissue Expression of PACAP, VIP, and Their Receptors in Response to Cold Stress. J Mol Neurosci 2018; 68:427-438. [PMID: 29982965 PMCID: PMC6581916 DOI: 10.1007/s12031-018-1099-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 06/07/2018] [Indexed: 12/15/2022]
Abstract
Obesity arises from disrupted energy balance and is caused by chronically higher energy intake compared to expenditure via basal metabolic rate, exercise, and thermogenesis. The brown adipose tissue (BAT), the primary thermogenic organ, has received considerable attention as a potential therapeutic target due to its ability to burn lipids in the production of heat. Pituitary adenylate cyclase-activating polypeptide (PACAP) has been identified as a key regulator of the physiological stress response both centrally and peripherally. While PACAP has been shown to increase thermogenesis by acting at the hypothalamus to increase sympathetic output to BAT, a peripheral role for PACAP-activated thermogenesis has not been studied. We identified PACAP receptor (PAC1, VPAC1/2) expression for the first time in murine BAT and confirmed their expression in white adipose tissues. PAC1 receptor expression was significantly altered in all three adipose tissues studied in response to 3.5-week cold acclimation, with expression patterns differing by depot type. In primary cell culture, VPAC1 was increased in differentiated compared to non-differentiated brown adipocytes, and the same trend was observed for the PACAP-specific receptor PAC1 in gonadal white fat primary cultures. The primary PAC1R mRNA splice variant in interscapular BAT was determined as isoform 2 by RNA-Seq. These results show that PACAP receptors are present in adipose tissues and may have important functional roles in adipocyte differentiation, lipid metabolism, or adipose sensitization to sympathetic signaling in response to thermogenic stimuli.
Collapse
|
29
|
Pituitary adenylate cyclase-activating polypeptide (PACAP) signaling in the prefrontal cortex modulates cued fear learning, but not spatial working memory, in female rats. Neuropharmacology 2018; 133:145-154. [DOI: 10.1016/j.neuropharm.2018.01.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 12/04/2017] [Accepted: 01/07/2018] [Indexed: 11/19/2022]
|
30
|
Fine morphological evaluation of hypothalamus in patients with hyperphagia. Acta Neurochir (Wien) 2017; 159:865-871. [PMID: 28224319 DOI: 10.1007/s00701-017-3112-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 02/01/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Various metabolic diseases induced by eating disorders are some of the most serious and difficult problems for modern public healthcare. However, little is known about hyperphagia, partly because of the lack of a clear definition. Several basic studies have analyzed eating habits using endocrinological or neurophysiological approaches, which have suggested a controlled balance between the hunger and satiety centers in the central nervous system. However, more detailed neuro-radiologic evaluations have not been achieved for the hypothalamus, and evaluations were limited only to the floor of the third ventricles. METHODS Fine structures of hypothalamic morphology were investigated using high-resolution magnetic resonance imaging in seven patients with hypothalamo-pituitary tumors, who suffered from preoperative hyperphagia-induced severe obesity and metabolic disorders. Body mass index (BMI) varied from 22.4 to 40.5 kg/m2 (mean 32.8 kg/m2). Clinical data were compared with the data of nine patients without hyperphagia and seven healthy volunteers. RESULTS Morphological evaluation was possible in all patients and control subjects, and patients with hyperphagia had significantly shortened maximum distances between the ependymal layers of the lateral wall of the third ventricle and fornixes (hyperphagia group right side 0.30 mm, left side 0.23 mm vs. patients without hyperphagia group right side 1.60, left side 1.53 vs. healthy group right side 1.73 mm, left side 1.85 mm) (p < 0.01). Two patients achieved postoperative improvement in both clinical and neuro-radiological findings. CONCLUSION Eating and metabolic disorders are related to strong dysfunction of the medial nuclei of the hypothalamus in patients with hypothalamo-pituitary tumors. We report the first case of dynamic improvement from hyperphagia, with both symptomatic and neuro-radiological findings.
Collapse
|
31
|
Sekar R, Wang L, Chow BKC. Central Control of Feeding Behavior by the Secretin, PACAP, and Glucagon Family of Peptides. Front Endocrinol (Lausanne) 2017; 8:18. [PMID: 28223965 PMCID: PMC5293785 DOI: 10.3389/fendo.2017.00018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/18/2017] [Indexed: 12/25/2022] Open
Abstract
Constituting a group of structurally related brain-gut peptides, secretin (SCT), pituitary adenylate cyclase-activating peptide (PACAP), and glucagon (GCG) family of peptide hormones exert their functions via interactions with the class B1 G protein-coupled receptors. In recent years, the roles of these peptides in neuroendocrine control of feeding behavior have been a specific area of research focus for development of potential therapeutic drug targets to combat obesity and metabolic disorders. As a result, some members in the family and their analogs have already been utilized as therapeutic agents in clinical application. This review aims to provide an overview of the current understanding on the important role of SCT, PACAP, and GCG family of peptides in central control of feeding behavior.
Collapse
Affiliation(s)
- Revathi Sekar
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Lei Wang
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | | |
Collapse
|
32
|
Wittmann G, Farkas E, Szilvásy-Szabó A, Gereben B, Fekete C, Lechan RM. Variable proopiomelanocortin expression in tanycytes of the adult rat hypothalamus and pituitary stalk. J Comp Neurol 2016; 525:411-441. [PMID: 27503597 DOI: 10.1002/cne.24090] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 08/01/2016] [Accepted: 08/04/2016] [Indexed: 01/21/2023]
Abstract
It is generally believed that proopiomelanocortin (POMC) is expressed exclusively by neurons in the adult rodent brain. Unbeknownst to most researchers, however, Pomc in situ hybridization studies in the rat show specific labeling in the ventral wall of the hypothalamic third ventricle, which is formed by specialized ependymal cells, called tanycytes. Here we characterized this non-neuronal POMC expression in detail using in situ hybridization and immunohistochemical techniques, and report two unique characteristics. First, POMC mRNA and precursor protein expression in non-neuronal cells varies to a great degree as to the extent and abundance of expression. In brains with low-level expression, POMC mRNA and protein was largely confined to a population of tanycytes within the infundibular stalk/caudal median eminence, termed here γ tanycytes, and a subset of closely located β and α2 tanycytes. In brains with high-level expression, POMC mRNA and protein was observed in the vast majority of α2, β, and γ tanycytes. This variability was observed in both adult males and females; of 41 rats between 8 and 15 weeks of age, 17 had low-, 9 intermediate-, and 15 high-level POMC expression in tanycytes. Second, unlike other known POMC-expressing cells, tanycytes rarely contained detectable levels of adrenocorticotropin or α-melanocyte-stimulating hormone. The results indicate either a dynamic spatiotemporal pattern whereby low and high POMC syntheses in tanycytes occur periodically in each brain, or marked interindividual differences that may persist throughout adulthood. Future studies are required to examine these possibilities and elucidate the physiologic importance of POMC in tanycytes. J. Comp. Neurol. 525:411-441, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Gábor Wittmann
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts, 02111
| | - Erzsébet Farkas
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, 1083, Hungary.,Pázmány Péter Catholic University, Multidisciplinary Doctoral School of Sciences and Technology, Budapest, 1083, Hungary
| | - Anett Szilvásy-Szabó
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, 1083, Hungary.,Semmelweis University, János Szentágothai PhD School of Neurosciences, Budapest, 1085, Hungary
| | - Balázs Gereben
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, 1083, Hungary
| | - Csaba Fekete
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts, 02111.,Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, 1083, Hungary
| | - Ronald M Lechan
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts, 02111.,Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111
| |
Collapse
|
33
|
Rudecki AP, Gray SL. PACAP in the Defense of Energy Homeostasis. Trends Endocrinol Metab 2016; 27:620-632. [PMID: 27166671 DOI: 10.1016/j.tem.2016.04.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/12/2016] [Accepted: 04/12/2016] [Indexed: 11/23/2022]
Abstract
The neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) mediates diverse physiology from neuroprotection to thermoregulation. PACAP is well established as a master regulator of the stress response, regulating psychological and physiological equilibrium via the autonomic nervous system. Neuroanatomical and functional evidence support a role for PACAP in energy metabolism, including thermogenesis, activity, mobilization of energy stores, and appetite. Through integration of this evidence we suggest PACAP be included in the growing list of neuropeptides that mediate energy homeostasis. Future work to uncover the intricacies of PACAP expression and the molecular pathways responsible for PACAP signaling may show potential for this neuropeptide as a therapeutic target as well as further elucidate the complex neuroanatomical networks involved in defending energy balance.
Collapse
Affiliation(s)
- Alexander P Rudecki
- Northern Medical Program, University of Northern British Columbia, 3333 University Way, Prince George BC, V2N 4Z9, Canada
| | - Sarah L Gray
- Northern Medical Program, University of Northern British Columbia, 3333 University Way, Prince George BC, V2N 4Z9, Canada.
| |
Collapse
|
34
|
Hurley MM, Maunze B, Block ME, Frenkel MM, Reilly MJ, Kim E, Chen Y, Li Y, Baker DA, Liu QS, Choi S. Pituitary Adenylate-Cyclase Activating Polypeptide Regulates Hunger- and Palatability-Induced Binge Eating. Front Neurosci 2016; 10:383. [PMID: 27597817 PMCID: PMC4993128 DOI: 10.3389/fnins.2016.00383] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/05/2016] [Indexed: 11/13/2022] Open
Abstract
While pituitary adenylate cyclase activating polypeptide (PACAP) signaling in the hypothalamic ventromedial nuclei (VMN) has been shown to regulate feeding, a challenge in unmasking a role for this peptide in obesity is that excess feeding can involve numerous mechanisms including homeostatic (hunger) and hedonic-related (palatability) drives. In these studies, we first isolated distinct feeding drives by developing a novel model of binge behavior in which homeostatic-driven feeding was temporally separated from feeding driven by food palatability. We found that stimulation of the VMN, achieved by local microinjections of AMPA, decreased standard chow consumption in food-restricted rats (e.g., homeostatic feeding); surprisingly, this manipulation failed to alter palatable food consumption in satiated rats (e.g., hedonic feeding). In contrast, inhibition of the nucleus accumbens (NAc), through local microinjections of GABA receptor agonists baclofen and muscimol, decreased hedonic feeding without altering homeostatic feeding. PACAP microinjections produced the site-specific changes in synaptic transmission needed to decrease feeding via VMN or NAc circuitry. PACAP into the NAc mimicked the actions of GABA agonists by reducing hedonic feeding without altering homeostatic feeding. In contrast, PACAP into the VMN mimicked the actions of AMPA by decreasing homeostatic feeding without affecting hedonic feeding. Slice electrophysiology recordings verified PACAP excitation of VMN neurons and inhibition of NAc neurons. These data suggest that the VMN and NAc regulate distinct circuits giving rise to unique feeding drives, but that both can be regulated by the neuropeptide PACAP to potentially curb excessive eating stemming from either drive.
Collapse
Affiliation(s)
- Matthew M Hurley
- Department of Biomedical Sciences, Marquette University Milwaukee, WI, USA
| | - Brian Maunze
- Department of Biomedical Sciences, Marquette University Milwaukee, WI, USA
| | - Megan E Block
- Department of Biomedical Sciences, Marquette University Milwaukee, WI, USA
| | - Mogen M Frenkel
- Department of Biomedical Sciences, Marquette University Milwaukee, WI, USA
| | - Michael J Reilly
- Department of Biomedical Sciences, Marquette University Milwaukee, WI, USA
| | - Eugene Kim
- Department of Biomedical Sciences, Marquette University Milwaukee, WI, USA
| | - Yao Chen
- Department of Pharmacology and Toxicology, Medical College of Wisconsin Milwaukee, WI, USA
| | - Yan Li
- Department of Pharmacology and Toxicology, Medical College of Wisconsin Milwaukee, WI, USA
| | - David A Baker
- Department of Biomedical Sciences, Marquette University Milwaukee, WI, USA
| | - Qing-Song Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin Milwaukee, WI, USA
| | - SuJean Choi
- Department of Biomedical Sciences, Marquette University Milwaukee, WI, USA
| |
Collapse
|
35
|
Iwasa T, Matsuzaki T, Tungalagsuvd A, Munkhzaya M, Yiliyasi M, Kato T, Kuwahara A, Irahara M. Developmental changes in the hypothalamic mRNA expression levels of PACAP and its receptor PAC1 and their sensitivity to fasting in male and female rats. Int J Dev Neurosci 2016; 52:33-7. [PMID: 27181029 DOI: 10.1016/j.ijdevneu.2016.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 04/28/2016] [Accepted: 05/11/2016] [Indexed: 01/02/2023] Open
Abstract
The actions and responses of hypothalamic appetite regulatory and factors change markedly during the neonatal to pre-pubertal period. Pituitary adenylate cyclase-activating polypeptide (PACAP) has been found to play pivotal roles in the regulation of metabolic and nutritional status through its specific receptor PAC1. PACAP/PAC1 have anorectic roles, and their functions are regulated by leptin in adulthood. In the present study, we showed that hypothalamic PACAP mRNA expression decreases during the neonatal to pre-pubertal period (from postnatal day 10-30) in both male and female rats. During this period, hypothalamic PACAP mRNA expression was not affected by 24h fasting in either sex, while the serum leptin levels (leptin is a positive regulator of hypothalamic PACAP expression in adulthood) of both sexes were decreased by fasting. On the other hand, hypothalamic PAC1 mRNA expression did not change during the neonatal to pre-pubertal period in either sex; however, its levels were consistently higher in males than in females. Hypothalamic PAC1 mRNA expression was decreased by 24h fasting in males, but no such changes were observed in females. These results indicate while hypothalamic PACAP expression is sensitive to a negative energy state and the serum leptin level in adulthood, no such relationships are seen in the pre-pubertal period. In addition, we speculate that differences in the gonadal steroidal milieu might induce sexual dimorphism in the basal hypothalamic PAC1 mRNA level and its response to fasting. The mechanisms responsible for and the physiological effects of such changes in hypothalamic PACAP and PAC1 expression during the developmental period remain to be clarified.
Collapse
Affiliation(s)
- Takeshi Iwasa
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-Cho, Tokushima 770-8503, Japan.
| | - Toshiya Matsuzaki
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Altankhuu Tungalagsuvd
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Munkhsaikhan Munkhzaya
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Maira Yiliyasi
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Takeshi Kato
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Akira Kuwahara
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-Cho, Tokushima 770-8503, Japan
| | - Minoru Irahara
- Department of Obstetrics and Gynecology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-Cho, Tokushima 770-8503, Japan
| |
Collapse
|
36
|
Ávila-Mendoza J, Carranza M, Villalobos P, Olvera A, Orozco A, Luna M, Arámburo C. Differential responses of the somatotropic and thyroid axes to environmental temperature changes in the green iguana. Gen Comp Endocrinol 2016; 230-231:76-86. [PMID: 27044512 DOI: 10.1016/j.ygcen.2016.04.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Revised: 03/23/2016] [Accepted: 04/01/2016] [Indexed: 11/26/2022]
Abstract
Growth hormone (GH), together with thyroid hormones (TH), regulates growth and development, and has critical effects on vertebrate metabolism. In ectotherms, these physiological processes are strongly influenced by environmental temperature. In reptiles, however, little is known about the direct influences of this factor on the somatotropic and thyroid axes. Therefore, the aim of this study was to describe the effects of both acute (48h) and chronic (2weeks) exposure to sub-optimal temperatures (25 and 18°C) upon somatotropic and thyroid axis function of the green iguana, in comparison to the control temperature (30-35°C). We found a significant increase in GH release (2.0-fold at 25°C and 1.9-fold at 18°C) and GH mRNA expression (up to 3.7-fold), mainly under chronic exposure conditions. The serum concentration of insulin-like growth factor-I (IGF-I) was significantly greater after chronic exposure (18.5±2.3 at 25°C; 15.92±3.4 at 18°C; vs. 9.3±1.21ng/ml at 35°C), while hepatic IGF-I mRNA expression increased up to 6.8-fold. Somatotropic axis may be regulated, under acute conditions, by thyrotropin-releasing hormone (TRH) that significantly increased its hypothalamic concentration (1.45 times) and mRNA expression (0.9-fold above control), respectively; and somatostatin (mRNA expression increased 1.0-1.2 times above control); and under chronic treatment, by pituitary adenylate cyclase-activating peptide (PACAP mRNA expression was increased from 0.4 to 0.6 times). Also, it was shown that, under control conditions, injection of TRH stimulated a significant increase in circulating GH. On the other hand, while there was a significant rise in the hypothalamic content of TRH and its mRNA expression, this hormone did not appear to influence the thyroid axis activity, which showed a severe diminution in all conditions of cold exposure, as indicated by the decreases in thyrotropin (TSH) mRNA expression (up to one-eight of the control), serum T4 (from 11.6±1.09 to 5.3±0.58ng/ml, after 2weeks at 18°C) and T3 (from 0.87±0.09 to 0.05±0.01ng/ml, under chronic conditions at 25°C), and Type-2 deiodinase (D2) activity (from 992.5±224 to 213.6±26.4fmolI(125)T4/mgh). The reduction in thyroid activity correlates with the down-regulation of metabolism as suggested by the decrease in the serum glucose and free fatty acid levels. These changes apparently were independent of a possible stress response, at least under acute exposure to both temperatures and in chronic treatment to 25°C, since serum corticosterone had no significant changes in these conditions, while at chronic 18°C exposure, a slight increase (0.38 times above control) was found. Thus, these data suggest that the reptilian somatotropic and thyroid axes have differential responses to cold exposure, and that GH and TRH may play important roles associated to adaptation mechanisms that support temperature acclimation in the green iguana.
Collapse
Affiliation(s)
- José Ávila-Mendoza
- Laboratorio de Bioquímica de Hormonas, Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, Qro. 76230, Mexico
| | - Martha Carranza
- Laboratorio de Bioquímica de Hormonas, Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, Qro. 76230, Mexico
| | - Patricia Villalobos
- Laboratorio de Fisiología Evolutiva, Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, Qro. 76230, Mexico
| | - Aurora Olvera
- Laboratorio de Fisiología Evolutiva, Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, Qro. 76230, Mexico
| | - Aurea Orozco
- Laboratorio de Fisiología Evolutiva, Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, Qro. 76230, Mexico
| | - Maricela Luna
- Laboratorio de Bioquímica de Hormonas, Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, Qro. 76230, Mexico
| | - Carlos Arámburo
- Laboratorio de Bioquímica de Hormonas, Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, Qro. 76230, Mexico.
| |
Collapse
|
37
|
Ueno H, Nakazato M. Mechanistic relationship between the vagal afferent pathway, central nervous system and peripheral organs in appetite regulation. J Diabetes Investig 2016; 7:812-818. [PMID: 27180615 PMCID: PMC5089941 DOI: 10.1111/jdi.12492] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 02/01/2016] [Accepted: 02/03/2016] [Indexed: 02/04/2023] Open
Abstract
The hypothalamus is a center of food intake and energy metabolism regulation. Information signals from peripheral organs are mediated through the circulation or the vagal afferent pathway and input into the hypothalamus, where signals are integrated to determine various behaviors, such as eating. Numerous appetite-regulating peptides are expressed in the central nervous system and the peripheral organs, and interact in a complex manner. Of such peptides, gut peptides are known to bind to receptors at the vagal afferent pathway terminal that extend into the mucosal layer of the digestive tract, modulate the electrical activity of the vagus nerve, and subsequently send signals to the solitary nucleus and furthermore to the hypothalamus. All peripheral peptides other than ghrelin suppress appetite, and they synergistically suppress appetite through the vagus nerve. In contrast, the appetite-enhancing peptide, ghrelin, antagonizes the actions of appetite-suppressing peptides through the vagus nerve, and appetite-suppressing peptides have attenuated effects in obesity as a result of inflammation in the vagus nerve. With greater understanding of the mechanism for food intake and energy metabolism regulation, medications that apply the effects of appetite-regulating peptides or implantable devices that electrically stimulate the vagus nerve are being investigated as novel treatments for obesity in basic and clinical studies.
Collapse
Affiliation(s)
- Hiroaki Ueno
- Division of Neurology, Respirology, Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Masamitsu Nakazato
- Division of Neurology, Respirology, Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan.
| |
Collapse
|
38
|
Vu JP, Goyal D, Luong L, Oh S, Sandhu R, Norris J, Parsons W, Pisegna JR, Germano PM. PACAP intraperitoneal treatment suppresses appetite and food intake via PAC1 receptor in mice by inhibiting ghrelin and increasing GLP-1 and leptin. Am J Physiol Gastrointest Liver Physiol 2015; 309:G816-25. [PMID: 26336928 PMCID: PMC4652141 DOI: 10.1152/ajpgi.00190.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 08/31/2015] [Indexed: 01/31/2023]
Abstract
Pituitary adenylate cyclase-activating peptide (PACAP) is expressed within the gastroenteric system, where it has profound physiological effects. PACAP was shown to regulate food intake and thermogenesis centrally; however, PACAP peripheral regulation of appetite and feeding behavior is unknown. Therefore, we studied PACAP's effect on appetite and food intake control by analyzing feeding behavior and metabolic hormones in PAC1-deficient (PAC1-/-) and age-matched wild-type (WT) mice intraperitoneally injected with PACAP1-38 or PACAP1-27 before the dark phase of feeding. Food intake and feeding behavior were analyzed using the BioDAQ system. Active ghrelin, glucagon-like peptide-1 (GLP-1), leptin, peptide YY, pancreatic polypeptide, and insulin were measured following PACAP1-38 administration in fasted WT mice. PACAP1-38/PACAP1-27 injected into WT mice significantly decreased in a dose-dependent manner cumulative food intake and reduced bout and meal feeding parameters. Conversely, PACAP1-38 injected into PAC1-/- mice failed to significantly change food intake. Importantly, PACAP1-38 reduced plasma levels of active ghrelin compared with vehicle in WT mice. In PAC1-/- mice, fasting levels of active ghrelin, GLP-1, insulin, and leptin and postprandial levels of active ghrelin and insulin were significantly altered compared with levels in WT mice. Therefore, PAC1 is a novel regulator of appetite/satiety. PACAP1-38/PACAP1-27 significantly reduced appetite and food intake through PAC1. In PAC1-/- mice, the regulation of anorexigenic/orexigenic hormones was abolished, whereas active ghrelin remained elevated even postprandially. PACAP significantly reduced active ghrelin in fasting conditions. These results establish a role for PACAP via PAC1 in the peripheral regulation of appetite/satiety and suggest future studies to explore a therapeutic use of PACAP or PAC1 agonists for obesity treatment.
Collapse
Affiliation(s)
- John P. Vu
- 1Research Service, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California;
| | - Deepinder Goyal
- 2Division of Digestive Diseases, David Geffen School of Medicine, University of California, Los Angeles, California;
| | - Leon Luong
- 3Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, California;
| | - Suwan Oh
- 1Research Service, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California;
| | - Ravneet Sandhu
- 1Research Service, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California;
| | - Joshua Norris
- 1Research Service, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California;
| | - William Parsons
- 1Research Service, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California; ,3Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, California;
| | - Joseph R. Pisegna
- 1Research Service, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California; ,3Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, California; ,4Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California; ,5CURE/Digestive Diseases Research Center, Department of Medicine, University of California, Los Angeles, California; and
| | - Patrizia M. Germano
- 1Research Service, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California; ,5CURE/Digestive Diseases Research Center, Department of Medicine, University of California, Los Angeles, California; and ,6Division of Pulmonary and Critical Care, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California
| |
Collapse
|
39
|
Seiglie MP, Smith KL, Blasio A, Cottone P, Sabino V. Pituitary adenylate cyclase-activating polypeptide induces a depressive-like phenotype in rats. Psychopharmacology (Berl) 2015; 232:3821-31. [PMID: 26264905 PMCID: PMC4565740 DOI: 10.1007/s00213-015-4045-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 07/30/2015] [Indexed: 12/27/2022]
Abstract
BACKGROUND Major depressive disorder (MDD) is a chronic, life-threatening psychiatric condition characterized by depressed mood, psychomotor alterations, and a markedly diminished interest or pleasure in most activities known as anhedonia. Available pharmacotherapies have limited success and the need for new strategies is clear. Recent studies attribute a major role to the pituitary adenylate cyclase-activating polypeptide (PACAP) system in mediating the response to stress. PACAP knockout mice display profound alterations in depressive-like behaviors, and genetic association studies have demonstrated that genetic variants of the PACAP gene are associated with MDD. However, the effects of PACAP administration on depressive-like behaviors in rodents have not yet been systematically examined. OBJECTIVES The present study investigated the effects of central administration of PACAP in rats on depressive-like behaviors, using well-established animal models that represent some of the endophenotypes of depression. METHODS We used intracranial self-stimulation (ICSS) to assess the brain reward function, saccharin preference test to assess anhedonia, social interaction to assess social withdrawal, and forced swim test (FST) to assess behavioral despair. RESULTS PACAP raised the current threshold for ICSS, elevation blocked by the PACAP antagonist PACAP(6-38). PACAP reduced the preference for a sweet saccharin solution and reduced the time the rats spent interacting with a novel animal. Interestingly, PACAP administration did not affect immobility in the FST. CONCLUSIONS Our results demonstrate a role for the central PACAP/PAC1R system in the regulation of depressive-like behaviors and suggest that hyperactivity of the PACAP/PAC1R system may contribute to the pathophysiology of depression, particularly the associated anhedonic symptomatology and social dysfunction.
Collapse
Affiliation(s)
- Mariel P. Seiglie
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA, Graduate Program in Neuroscience, Boston University School of Medicine, Boston, MA
| | - Karen L. Smith
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA
| | - Angelo Blasio
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA
| | - Pietro Cottone
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA
| | - Valentina Sabino
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University School of Medicine, Boston, MA
| |
Collapse
|
40
|
Hammack SE, May V. Pituitary adenylate cyclase activating polypeptide in stress-related disorders: data convergence from animal and human studies. Biol Psychiatry 2015; 78:167-77. [PMID: 25636177 PMCID: PMC4461555 DOI: 10.1016/j.biopsych.2014.12.003] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 11/13/2014] [Accepted: 12/01/2014] [Indexed: 12/21/2022]
Abstract
The maladaptive expression and function of several stress-associated hormones have been implicated in pathological stress and anxiety-related disorders. Among these, recent evidence has suggested that pituitary adenylate cyclase activating polypeptide (PACAP) has critical roles in central neurocircuits mediating stress-related emotional behaviors. We describe the PACAPergic systems, the data implicating PACAP in stress biology, and how altered PACAP expression and signaling may result in psychopathologies. We include our work implicating PACAP signaling within the bed nucleus of the stria terminalis in mediating the consequences of stressor exposure and relatedly, describe more recent studies suggesting that PACAP in the central nucleus of the amygdala may impact the emotional aspects of chronic pain states. In aggregate, these results are consistent with data suggesting that PACAP dysregulation is associated with posttraumatic stress disorder in humans.
Collapse
Affiliation(s)
- Sayamwong E. Hammack
- Department of Psychological Science, University of Vermont, John Dewey Hall, 2 Colchester Avenue, Burlington, Vermont 05405-0134, Phone: 802.656.1041, Fax: 802.656.8783
| | - Victor May
- Department of Neurological Sciences, University of Vermont College of Medicine, 149 Beaumont Avenue, HSRF 428, Burlington, VT 05405, Phone: 802.656.4579
| |
Collapse
|
41
|
Iemolo A, Ferragud A, Cottone P, Sabino V. Pituitary Adenylate Cyclase-Activating Peptide in the Central Amygdala Causes Anorexia and Body Weight Loss via the Melanocortin and the TrkB Systems. Neuropsychopharmacology 2015; 40:1846-55. [PMID: 25649277 PMCID: PMC4839508 DOI: 10.1038/npp.2015.34] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 01/10/2015] [Accepted: 01/11/2015] [Indexed: 02/03/2023]
Abstract
Growing evidence suggests that the pituitary adenylate cyclase-activating polypeptide (PACAP)/PAC1 receptor system represents one of the main regulators of the behavioral, endocrine, and autonomic responses to stress. Although induction of anorexia is a well-documented effect of PACAP, the central sites underlying this phenomenon are poorly understood. The present studies addressed this question by examining the neuroanatomical, behavioral, and pharmacological mechanisms mediating the anorexia produced by PACAP in the central nucleus of the amygdala (CeA), a limbic structure implicated in the emotional components of ingestive behavior. Male rats were microinfused with PACAP (0-1 μg per rat) into the CeA and home-cage food intake, body weight change, microstructural analysis of food intake, and locomotor activity were assessed. Intra-CeA (but not intra-basolateral amygdala) PACAP dose-dependently induced anorexia and body weight loss without affecting locomotor activity. PACAP-treated rats ate smaller meals of normal duration, revealing that PACAP slowed feeding within meals by decreasing the regularity and maintenance of feeding from pellet-to-pellet; postprandial satiety was unaffected. Intra-CeA PACAP-induced anorexia was blocked by coinfusion of either the melanocortin receptor 3/4 antagonist SHU 9119 or the tyrosine kinase B (TrKB) inhibitor k-252a, but not the CRF receptor antagonist D-Phe-CRF(12-41). These results indicate that the CeA is one of the brain areas through which the PACAP system promotes anorexia and that PACAP preferentially lessens the maintenance of feeding in rats, effects opposite to those of palatable food. We also demonstrate that PACAP in the CeA exerts its anorectic effects via local melanocortin and the TrKB systems, and independently from CRF.
Collapse
Affiliation(s)
- Attilio Iemolo
- Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA,Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Antonio Ferragud
- Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA,Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Pietro Cottone
- Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA,Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA,Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118 USA, Tel: +1 617 638 4327, Fax: +1 617 638 5668, E-mail: or
| | - Valentina Sabino
- Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA,Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA,Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118 USA, Tel: +1 617 638 4327, Fax: +1 617 638 5668, E-mail: or
| |
Collapse
|
42
|
Yu R, Yang Y, Cui Z, Zheng L, Zeng Z, Zhang H. Novel peptide VIP-TAT with higher affinity for PAC1 inhibited scopolamine induced amnesia. Peptides 2014; 60:41-50. [PMID: 25086267 DOI: 10.1016/j.peptides.2014.07.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Revised: 07/19/2014] [Accepted: 07/21/2014] [Indexed: 12/25/2022]
Abstract
A novel peptide VIP-TAT with a cell penetrating peptide TAT at the C-terminus of VIP was constructed and prepared using intein mediated purification with an affinity chitin-binding tag (IMPACT) system to enhance the brain uptake efficiency for the medical application in central nervous system. It was found by labeling VIP-TAT and VIP with fluorescein isothiocyanate (FITC) that the extension with TAT increased the brain uptake efficiency of VIP-TAT significantly. Then short-term and long-term treatment with scopolamine (Scop) was used to evaluate the effect of VIP-TAT or VIP on Scop induced amnesia. Both short-term and long-term administration of VIP-TAT inhibited the latent time reduction in step-through test induced by Scop significantly, but long-term administration of VIP aggravated the Scop induced amnesia. Long-term i.p. injection of VIP-TAT was shown to have positive effect by inhibiting the oxidative damage, apoptosis and the cholinergic system activity reduction that induced by Scop, while VIP exerted negative effect in brain opposite to that in periphery system. The in vitro data showed that VIP-TAT had not only protective but also proliferative effect on Neuro2a cells which was inhibited by PAC1 antagonist PACAP(6-38). Competition binding assay and cAMP assay confirmed that VIP-TAT had higher affinity and activation for PAC1 than VIP. So it was concluded that the significantly stronger protective effect of VIP-TAT against Scop induced amnesia than VIP was due to (1) the enhanced brain uptake efficiency of VIP-TAT and (2) the increased affinity and activation of VIP-TAT for receptor PAC1.
Collapse
Affiliation(s)
- Rongjie Yu
- Cell Biology Institute, Department of Cell Biology, Jinan University, Guangzhou 510632, China.
| | - Yanxu Yang
- Cell Biology Institute, Department of Cell Biology, Jinan University, Guangzhou 510632, China
| | - Zekai Cui
- Cell Biology Institute, Department of Cell Biology, Jinan University, Guangzhou 510632, China
| | - Lijun Zheng
- Cell Biology Institute, Department of Cell Biology, Jinan University, Guangzhou 510632, China
| | - Zhixing Zeng
- Cell Biology Institute, Department of Cell Biology, Jinan University, Guangzhou 510632, China
| | - Huahua Zhang
- Laboratory of Medical Genetics of Guangdong Medical College, Dongguan, Guangdong 523808, China
| |
Collapse
|
43
|
Banki E, Pakai E, Gaszner B, Zsiboras C, Czett A, Bhuddi PRP, Hashimoto H, Toth G, Tamas A, Reglodi D, Garami A. Characterization of the thermoregulatory response to pituitary adenylate cyclase-activating polypeptide in rodents. J Mol Neurosci 2014; 54:543-54. [PMID: 24994541 DOI: 10.1007/s12031-014-0361-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 06/23/2014] [Indexed: 12/19/2022]
Abstract
Administration of the long form (38 amino acids) of pituitary adenylate cyclase-activating polypeptide (PACAP38) into the central nervous system causes hyperthermia, suggesting that PACAP38 plays a role in the regulation of deep body temperature (T b). In this study, we investigated the thermoregulatory role of PACAP38 in details. First, we infused PACAP38 intracerebroventricularly to rats and measured their T b and autonomic thermoeffector responses. We found that central PACAP38 infusion caused dose-dependent hyperthermia, which was brought about by increased thermogenesis and tail skin vasoconstriction. Compared to intracerebroventricular administration, systemic (intravenous) infusion of the same dose of PACAP38 caused significantly smaller hyperthermia, indicating a central site of action. We then investigated the thermoregulatory phenotype of mice lacking the Pacap gene (Pacap (-/-)). Freely moving Pacap (-/-) mice had higher locomotor activity throughout the day and elevated deep T b during the light phase. When the Pacap (-/-) mice were loosely restrained, their metabolic rate and T b were lower compared to their wild-type littermates. We conclude that PACAP38 causes hyperthermia via activation of the autonomic cold-defense thermoeffectors through central targets. Pacap (-/-) mice express hyperkinesis, which is presumably a compensatory mechanism, because under restrained conditions, these mice are hypometabolic and hypothermic compared to controls.
Collapse
Affiliation(s)
- Eszter Banki
- Department of Anatomy PTE-MTA "Lendulet" PACAP Research Team, Medical School, University of Pecs, Pecs, Hungary
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Resch JM, Maunze B, Phillips KA, Choi S. Inhibition of food intake by PACAP in the hypothalamic ventromedial nuclei is mediated by NMDA receptors. Physiol Behav 2014; 133:230-5. [PMID: 24878316 DOI: 10.1016/j.physbeh.2014.05.029] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 05/02/2014] [Accepted: 05/07/2014] [Indexed: 11/17/2022]
Abstract
Central injections of pituitary adenylate cyclase-activating polypeptide (PACAP) into the ventromedial nuclei (VMN) of the hypothalamus produce hypophagia that is dependent upon the PAC1 receptor; however, the signaling downstream of this receptor in the VMN is unknown. Though PACAP signaling has many targets, this neuropeptide has been shown to influence glutamate signaling in several brain regions through mechanisms involving NMDA receptor potentiation via activation of the Src family of protein tyrosine kinases. With this in mind, we examined the Src-NMDA receptor signaling pathway as a target for PACAP signaling in the VMN that may mediate its effects on feeding behavior. Under nocturnal feeding conditions, NMDA receptor antagonism prior to PACAP administration into the VMN attenuated PACAP-mediated decreases in feeding suggesting that glutamatergic signaling via NMDA receptors is necessary for PACAP-induced hypophagia. Furthermore, PACAP administration into the VMN resulted in increased tyrosine phosphorylation of the GluN2B subunit of the NMDA receptor, and inhibition of Src kinase activity also blocked the effects of PACAP administration into the VMN on feeding behavior. These results indicate that PACAP neurotransmission in the VMN likely augments glutamate signaling by potentiating NMDA receptors activity through the tyrosine phosphorylation events mediated by the Src kinase family, and modulation of NMDA receptor activity by PACAP in the hypothalamus may be a primary mechanism for its regulation of food intake.
Collapse
Affiliation(s)
- Jon M Resch
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI 53201, USA
| | - Brian Maunze
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI 53201, USA
| | - Kailynn A Phillips
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI 53201, USA
| | - SuJean Choi
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI 53201, USA.
| |
Collapse
|
45
|
Köves K, Kántor O, Lakatos A, Szabó E, Kirilly E, Heinzlmann A, Szabó F. Advent and recent advances in research on the role of pituitary adenylate cyclase-activating polypeptide (PACAP) in the regulation of gonadotropic hormone secretion of female rats. J Mol Neurosci 2014; 54:494-511. [PMID: 24696167 DOI: 10.1007/s12031-014-0294-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 03/20/2014] [Indexed: 02/06/2023]
Abstract
PACAP (ADCYAP1) was isolated from ovine hypothalami. PACAP activates three distinct receptor types: G-protein coupled PAC1, VPAC1, and VPAC2 with seven transmembrane domains. Eight splice variants of PAC1 receptor are described. A part of the hypothalamic PACAP is released into the hypophyseal portal circulation. Both hypothalamic and pituitary PACAP are involved in the dynamic control of gonadotropic hormone secretion. In female rats, PACAP in the paraventricular nucleus is upregulated in the morning and pituitary PACAP is upregulated in the late evening of the proestrus stage of the reproductive cycle. PACAP mRNA peak in the hypothalamic PVN precedes the LHRH release into the portal circulation. It is supposed that PACAP peak is evoked by the elevated estrogen on proestrous morning. At the beginning of the so-called critical period of the same day, PACAP level starts to decline allowing LHRH release into the portal circulation, resulting in the LH surge that evokes ovulation. Just before the critical period, icv-administered exogenous PACAP blocks the LH surge and ovulation. The blocking effect of PACAP is mediated through CRF and endogenous opioids. The effect of the pituitary-born PACAP depends on the intracellular cross-talk between PACAP and LHRH.
Collapse
Affiliation(s)
- Katalin Köves
- Department of Human Morphology and Developmental Biology, Faculty of Medicine, Semmelweis University, Tűzoltó u. 58, H-1094, Budapest, Hungary,
| | | | | | | | | | | | | |
Collapse
|
46
|
Khan AM. Controlling feeding behavior by chemical or gene-directed targeting in the brain: what's so spatial about our methods? Front Neurosci 2013; 7:182. [PMID: 24385950 PMCID: PMC3866545 DOI: 10.3389/fnins.2013.00182] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 09/20/2013] [Indexed: 12/26/2022] Open
Abstract
Intracranial chemical injection (ICI) methods have been used to identify the locations in the brain where feeding behavior can be controlled acutely. Scientists conducting ICI studies often document their injection site locations, thereby leaving kernels of valuable location data for others to use to further characterize feeding control circuits. Unfortunately, this rich dataset has not yet been formally contextualized with other published neuroanatomical data. In particular, axonal tracing studies have delineated several neural circuits originating in the same areas where ICI injection feeding-control sites have been documented, but it remains unclear whether these circuits participate in feeding control. Comparing injection sites with other types of location data would require careful anatomical registration between the datasets. Here, a conceptual framework is presented for how such anatomical registration efforts can be performed. For example, by using a simple atlas alignment tool, a hypothalamic locus sensitive to the orexigenic effects of neuropeptide Y (NPY) can be aligned accurately with the locations of neurons labeled by anterograde tracers or those known to express NPY receptors or feeding-related peptides. This approach can also be applied to those intracranial "gene-directed" injection (IGI) methods (e.g., site-specific recombinase methods, RNA expression or interference, optogenetics, and pharmacosynthetics) that involve viral injections to targeted neuronal populations. Spatial alignment efforts can be accelerated if location data from ICI/IGI methods are mapped to stereotaxic brain atlases to allow powerful neuroinformatics tools to overlay different types of data in the same reference space. Atlas-based mapping will be critical for community-based sharing of location data for feeding control circuits, and will accelerate our understanding of structure-function relationships in the brain for mammalian models of obesity and metabolic disorders.
Collapse
Affiliation(s)
- Arshad M. Khan
- UTEP Systems Neuroscience Laboratory, Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El PasoEl Paso, TX, USA
- Neurobiology Section, Department of Biological Sciences, University of Southern CaliforniaLos Angeles, CA, USA
| |
Collapse
|
47
|
Resch JM, Maunze B, Gerhardt AK, Magnuson SK, Phillips KA, Choi S. Intrahypothalamic pituitary adenylate cyclase-activating polypeptide regulates energy balance via site-specific actions on feeding and metabolism. Am J Physiol Endocrinol Metab 2013; 305:E1452-63. [PMID: 24148346 PMCID: PMC3882380 DOI: 10.1152/ajpendo.00293.2013] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Numerous studies have demonstrated that both the hypothalamic paraventricular nuclei (PVN) and ventromedial nuclei (VMN) regulate energy homeostasis through behavioral and metabolic mechanisms. Receptors for pituitary adenylate cyclase-activating polypeptide (PACAP) are abundantly expressed in these nuclei, suggesting PACAP may be critical for the regulation of feeding behavior and body weight. To characterize the unique behavioral and physiological responses attributed to select hypothalamic cell groups, PACAP was site-specifically injected into the PVN or VMN. Overall food intake was significantly reduced by PACAP at both sites; however, meal pattern analysis revealed that only injections into the PVN produced significant reductions in meal size, duration, and total time spent eating. PACAP-mediated hypophagia in both the PVN and VMN was abolished by PAC1R antagonism, whereas pretreatment with a VPACR antagonist had no effect. PACAP injections into the VMN produced unique changes in metabolic parameters, including significant increases in core body temperature and spontaneous locomotor activity that was PAC1R dependent whereas, PVN injections of PACAP had no effect. Finally, PACAP-containing afferents were identified using the neuronal tracer cholera toxin subunit B (CTB) injected unilaterally into the PVN or VMN. CTB signal from PVN injections was colocalized with PACAP mRNA in the medial anterior bed nucleus of the stria terminalis, VMN, and lateral parabrachial nucleus (LPB), whereas CTB signal from VMN injections was highly colocalized with PACAP mRNA in the medial amygdala and LPB. These brain regions are known to influence energy homeostasis perhaps, in part, through PACAP projections to the PVN and VMN.
Collapse
Affiliation(s)
- Jon M Resch
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin
| | | | | | | | | | | |
Collapse
|
48
|
Fokidis HB, Prior NH, Soma KK. Fasting increases aggression and differentially modulates local and systemic steroid levels in male zebra finches. Endocrinology 2013; 154:4328-39. [PMID: 23939990 DOI: 10.1210/en.2013-1171] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Aggression enables individuals to obtain and retain limited resources. Studies of the neuroendocrine regulation of aggression have focused on territorial and reproductive contexts. By contrast, little is understood concerning the neuroendocrine regulation of aggression over other resources, such as food. Here, we developed a paradigm to examine the role of steroids in food-related aggression. In groups of male zebra finches, a 6-hour fast decreased body mass and increased aggressive interactions among subjects that competed for a point source feeder. Fasting also dramatically altered circulating steroid levels by decreasing plasma testosterone but not estradiol (E2). By contrast, both plasma corticosterone and dehydroepiandrosterone (DHEA) concentrations were elevated with fasting. Interestingly, short-term access to food (15 minutes) after fasting normalized circulating steroid levels. Fasting increased corticosterone levels in a wide range of peripheral tissues but increased DHEA levels specifically in adrenal glands and liver; these effects were quickly normalized with refeeding. DHEA can be metabolized within specific brain regions to testosterone and E2, which promote the expression of aggression. We measured E2 in microdissected brain regions and found that fasting specifically increased local E2 levels in 3 regions: the periaqueductal gray, ventral tegmental area, and ventromedial nucleus of the hypothalamus. These regions are part of the vertebrate social behavior network and regulate the expression of aggression. Together, these data suggest that fasting stimulates secretion of DHEA from the adrenals and liver and subsequent conversion of DHEA to E2 within specific brain regions, to enable individuals to compete for limited food resources.
Collapse
Affiliation(s)
- H Bobby Fokidis
- Rollins College, 1000 Holt Avenue, Winter Park, Florida 32789-4499, USA
| | | | | |
Collapse
|
49
|
CRF mediates the anxiogenic and anti-rewarding, but not the anorectic effects of PACAP. Neuropsychopharmacology 2013; 38:2160-9. [PMID: 23657440 PMCID: PMC3773665 DOI: 10.1038/npp.2013.113] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 05/03/2013] [Accepted: 05/03/2013] [Indexed: 01/23/2023]
Abstract
Anxiety disorders represent the most common mental disturbances in the world, and they are characterized by an abnormal response to stress. Pituitary adenylate cyclase-activating polypeptide (PACAP) and its receptor PAC1 have been proposed to have a key role in mediating the responses to stress as well as the regulation of food intake and body weight. Corticotropin-releasing factor (CRF), the major stress peptide in the brain, has been hypothesized to be involved in PACAP effects, but the reports are conflicting so far. The present study was aimed at further characterizing the behavioral effects of PACAP in rats and at determining the role of central CRF receptors. We found that intracerebroventricular PACAP treatment induced anxiety-like behavior in the elevated plus maze test and elevated intracranial self-stimulation thresholds; both of these effects were fully blocked by concurrent treatment with the CRF receptor antagonist D-Phe-CRF(12-41). Interestingly, the CRF antagonist had no effect on PACAP-induced increased plasma corticosterone, reduction of food intake, and body weight loss. Finally, we found that PACAP increased CRF levels in the paraventricular nucleus of the hypothalamus and, importantly, in the central nucleus of the amygdala, as measured by solid phase radioimmunoassay and quantitative real-time PCR. Our results strengthen the notion that PACAP is a strong mediator of the behavioral response to stress and prove for the first time that this neuropeptide has anti-rewarding (ie, pro-depressant) effects. In addition, we identified the mechanism by which PACAP exerts its anxiogenic and pro-depressant effects, via the recruitment of the central CRF system and independently from HPA axis activation.
Collapse
|
50
|
Pituitary adenylate cyclase-activating polypeptide 6-38 blocks cocaine- and amphetamine-regulated transcript Peptide-induced hypophagia in rats. PLoS One 2013; 8:e72347. [PMID: 23967296 PMCID: PMC3744533 DOI: 10.1371/journal.pone.0072347] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 07/08/2013] [Indexed: 02/01/2023] Open
Abstract
Cocaine- and amphetamine-regulated transcript peptides (CARTp) suppress nutritional intake after administration into the fourth intracerebral ventricle. Recent in vitro studies have shown that PACAP 6-38, a pituitary adenylate cyclase-activating polypeptide (PACAP) fragment, could act as a competitive antagonist against CARTp 55-102 on a common CARTp-sensitive receptor structure. Here, we show for the first time in vivo that the reduction in solid food intake induced by exogenous CARTp 55-102 (0.3 nmol: 1.5 µg) administered fourth i.c.v. is blocked by pretreatment with PACAP 6-38 (3 nmol). The PACAP 6-38 fragment had no effect by itself either when given into the fourth ventricle or subcutaneously. Although effective to block the CARTp-effect on feeding and short-term body weight, PACAP 6-38 failed to attenuate CARTp-associated gross motor behavioral changes suggesting at least two CARTp-sensitive receptor subtypes. In conclusion, PACAP 6-38 acts as a functional CARTp antagonist in vivo and blocks its effects on feeding and short term weight gain.
Collapse
|