1
|
Magnan C, Migrenne-Li S. Pleiotropic effects of prokineticin 2 in the control of energy metabolism. Biochimie 2021; 186:73-81. [PMID: 33932486 DOI: 10.1016/j.biochi.2021.04.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/09/2021] [Accepted: 04/24/2021] [Indexed: 11/19/2022]
Abstract
Prokineticins are family of small proteins involved in many important biological processes including food intake and control of energy balance. The prokineticin 2 (PROK2) is expressed in several peripheral tissues and areas in the central nervous system. PROK2 activates G protein-coupled receptors, namely, prokineticin receptor 1 (PROKR1) and prokineticin receptor 2 (PROKR2). Preclinical models exhibiting disturbances of the PROK2 pathway (at the level of PROK2 or its receptors) are characterized by changes in food intake, feeding behavior and insulin sensitivity related to a dysfunction of the energy balance control. In Humans, mutations of PROK2 and PROKR2 genes are associated to the Kallmann syndrome (KS) that affects both the hormonal reproductive axis and the sense of smell and may also lead to obesity. Moreover, plasma PROK2 concentration has been correlated with various cardiometabolic risk factors and type 2 diabetes (T2D). The present review summarizes knowledge on PROK2 structure, signaling and function focusing on its role in control of food intake and energy homeostasis.
Collapse
|
2
|
Désaubry L, Kanthasamy AG, Nebigil CG. Prokineticin signaling in heart-brain developmental axis: Therapeutic options for heart and brain injuries. Pharmacol Res 2020; 160:105190. [PMID: 32937177 PMCID: PMC7674124 DOI: 10.1016/j.phrs.2020.105190] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/27/2020] [Accepted: 08/31/2020] [Indexed: 02/07/2023]
Abstract
Heart and brain development occur simultaneously during the embryogenesis, and both organ development and injuries are interconnected. Early neuronal and cardiac injuries share mutual cellular events, such as angiogenesis and plasticity that could either delay disease progression or, in the long run, result in detrimental health effects. For this reason, the common mechanisms provide a new and previously undervalued window of opportunity for intervention. Because angiogenesis, cardiogenesis and neurogenesis are essential for the development and regeneration of the heart and brain, we discuss therein the role of prokineticin as an angiogenic neuropeptide in heart-brain development and injuries. We focus on the role of prokineticin signaling and the effect of drugs targeting prokineticin receptors in neuroprotection and cardioprotection, with a special emphasis on heart failure, neurodegenerativParkinson's disease and ischemic heart and brain injuries. Indeed, prokineticin triggers common pro-survival signaling pathway in heart and brain. Our review aims at stimulating researchers and clinicians in neurocardiology to focus on the role of prokineticin signaling in the reciprocal interaction between heart and brain. We hope to facilitate the discovery of new treatment strategies, acting in both heart and brain degenerative diseases.
Collapse
Affiliation(s)
- Laurent Désaubry
- Regenerative Nanomedicine, UMR 1260, INSERM, University of Strasbourg, Strasbourg, France
| | - Anumantha G Kanthasamy
- Parkinson's Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, Iowa, USA
| | - Canan G Nebigil
- Regenerative Nanomedicine, UMR 1260, INSERM, University of Strasbourg, Strasbourg, France.
| |
Collapse
|
3
|
Northeast RC, Chrobok L, Hughes ATL, Petit C, Piggins HD. Keeping time in the lamina terminalis: Novel oscillator properties of forebrain sensory circumventricular organs. FASEB J 2019; 34:974-987. [PMID: 31914667 PMCID: PMC6972491 DOI: 10.1096/fj.201901111r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 09/19/2019] [Accepted: 10/15/2019] [Indexed: 12/11/2022]
Abstract
Drinking behavior and osmotic regulatory mechanisms exhibit clear daily variation which is necessary for achieving the homeostatic osmolality. In mammals, the master clock in the brain's suprachiasmatic nuclei has long been held as the main driver of circadian (24 h) rhythms in physiology and behavior. However, rhythmic clock gene expression in other brain sites raises the possibility of local circadian control of neural activity and function. The subfornical organ (SFO) and the organum vasculosum laminae terminalis (OVLT) are two sensory circumventricular organs (sCVOs) that play key roles in the central control of thirst and water homeostasis, but the extent to which they are subject to intrinsic circadian control remains undefined. Using a combination of ex vivo bioluminescence and in vivo gene expression, we report for the first time that the SFO contains an unexpectedly robust autonomous clock with unusual spatiotemporal characteristics in core and noncore clock gene expression. Furthermore, putative single-cell oscillators in the SFO and OVLT are strongly rhythmic and require action potential-dependent communication to maintain synchrony. Our results reveal that these thirst-controlling sCVOs possess intrinsic circadian timekeeping properties and raise the possibility that these contribute to daily regulation of drinking behavior.
Collapse
Affiliation(s)
- Rebecca C Northeast
- Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, UK
| | - Lukasz Chrobok
- Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, UK
| | - Alun T L Hughes
- Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, UK
| | - Cheryl Petit
- Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, UK
| | - Hugh D Piggins
- Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, UK
| |
Collapse
|
4
|
Negri L, Ferrara N. The Prokineticins: Neuromodulators and Mediators of Inflammation and Myeloid Cell-Dependent Angiogenesis. Physiol Rev 2018. [PMID: 29537336 DOI: 10.1152/physrev.00012.2017] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The mammalian prokineticins family comprises two conserved proteins, EG-VEGF/PROK1 and Bv8/PROK2, and their two highly related G protein-coupled receptors, PKR1 and PKR2. This signaling system has been linked to several important biological functions, including gastrointestinal tract motility, regulation of circadian rhythms, neurogenesis, angiogenesis and cancer progression, hematopoiesis, and nociception. Mutations in PKR2 or Bv8/PROK2 have been associated with Kallmann syndrome, a developmental disorder characterized by defective olfactory bulb neurogenesis, impaired development of gonadotropin-releasing hormone neurons, and infertility. Also, Bv8/PROK2 is strongly upregulated in neutrophils and other inflammatory cells in response to granulocyte-colony stimulating factor or other myeloid growth factors and functions as a pronociceptive mediator in inflamed tissues as well as a regulator of myeloid cell-dependent tumor angiogenesis. Bv8/PROK2 has been also implicated in neuropathic pain. Anti-Bv8/PROK2 antibodies or small molecule PKR inhibitors ameliorate pain arising from tissue injury and inhibit angiogenesis and inflammation associated with tumors or some autoimmune disorders.
Collapse
Affiliation(s)
- Lucia Negri
- Sapienza University of Rome, Rome, Italy ; and University of California, San Diego, La Jolla, California
| | - Napoleone Ferrara
- Sapienza University of Rome, Rome, Italy ; and University of California, San Diego, La Jolla, California
| |
Collapse
|
5
|
Zhao Y, Wu J, Wang X, Jia H, Chen DN, Li JD. Prokineticins and their G protein-coupled receptors in health and disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 161:149-179. [PMID: 30711026 DOI: 10.1016/bs.pmbts.2018.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prokineticins are two conserved small proteins (~8kDa), prokineticin 1 (PROK1; also called EG-VEGF) and prokineticin 2 (PROK2; also called Bv8), with an N-terminal AVITGA sequence and 10 cysteines forming 5 disulfide bridges. PROK1 and PROK2 bind to two highly related G protein-coupled receptors (GPCRs), prokineticin receptor 1 (PROKR1) and prokineticin receptor 2 (PROKR2). Prokineticins and their receptors are widely expressed. PROK1 is predominantly expressed in peripheral tissues, especially steroidogenic organs, whereas PROK2 is mainly expressed in the central nervous system and nonsteroidogenic cells of the testes. Prokineticins signaling has been implicated in several important physiological functions, including gastrointestinal smooth muscle contraction, circadian rhythm regulation, neurogenesis, angiogenesis, pain perception, mood regulation, and reproduction. Dysregulation of prokineticins signaling has been observed in a variety of diseases, such as cancer, ischemia, and neurodegeneration, in which prokineticins signaling seems to be a promising therapeutic target. Based on the phenotypes of knockout mice, PROKR2 and PROK2 have recently been identified as causative genes for idiopathic hypogonadotropic hypogonadism, a developmental disorder characterized by impaired development of gonadotropin-releasing hormone neurons and infertility. In vitro functional studies with these disease-associated PROKR2 mutations uncovered some novel features for this receptor, such as biased signaling, which may be used to understand GPCR signaling regulation in general.
Collapse
Affiliation(s)
- Yaguang Zhao
- School of Life Sciences, Central South University, Changsha, China; Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, China; Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, China
| | - Jiayu Wu
- School of Life Sciences, Central South University, Changsha, China; Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, China; Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, China
| | - Xinying Wang
- School of Life Sciences, Central South University, Changsha, China; Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, China; Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, China
| | - Hong Jia
- School of Life Sciences, Central South University, Changsha, China; Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, China; Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, China
| | - Dan-Na Chen
- Department of Basic Medical Sciences, Changsha Medical University, Changsha, China.
| | - Jia-Da Li
- School of Life Sciences, Central South University, Changsha, China; Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, China; Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, China.
| |
Collapse
|
6
|
Caioli S, Severini C, Ciotti T, Florenzano F, Pimpinella D, Petrocchi Passeri P, Balboni G, Polisca P, Lattanzi R, Nisticò R, Negri L, Zona C. Prokineticin system modulation as a new target to counteract the amyloid beta toxicity induced by glutamatergic alterations in an in vitro model of Alzheimer's disease. Neuropharmacology 2017; 116:82-97. [DOI: 10.1016/j.neuropharm.2016.12.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 11/29/2016] [Accepted: 12/14/2016] [Indexed: 12/28/2022]
|
7
|
Rhythmic Trafficking of TRPV2 in the Suprachiasmatic Nucleus is Regulated by Prokineticin 2 Signaling. J Circadian Rhythms 2015; 13:2. [PMID: 27103928 PMCID: PMC4832818 DOI: 10.5334/jcr.ad] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The mammalian circadian clock is composed of single-cell oscillators. Neurochemical and
electrical signaling among these oscillators is important for the normal expression of circadian
rhythms. Prokineticin 2 (PK2), encoding a cysteine-rich secreted protein, has been shown to be a
critical signaling molecule for the regulation of circadian rhythms. PK2 expression in the
suprachiasmatic nucleus (SCN) is highly rhythmic, peaking during the day and being essentially
absent during the night. Mice with disrupted PK2 gene or its receptor PKR2 display greatly reduced
rhythmicity of broad circadian parameters such as locomotor activity, body temperature and
sleep/wake patterns. PK2 has been shown to increase the firing rate of SCN neurons, with unknown
molecular mechanisms. Here we report that TRPV2, an ion channel belonging to the family of TRP, is
co-expressed with PKR2 in the SCN neurons. Further, TRPV2 protein, but not TRPV2 mRNA, was shown to
oscillate in the SCN in a PK2-dependent manner. Functional studies revealed that TRPV2 enhanced
signaling of PKR2 in calcium mobilization or ion current conductance, likely via the increased
trafficking of TRPV2 to the cell surface. Taken together, these results indicate that TRPV2 is
likely part of the downstream signaling of PK2 in the regulation of the circadian rhythms.
Collapse
|
8
|
Sakhi K, Wegner S, Belle MDC, Howarth M, Delagrange P, Brown TM, Piggins HD. Intrinsic and extrinsic cues regulate the daily profile of mouse lateral habenula neuronal activity. J Physiol 2014; 592:5025-45. [PMID: 25194046 DOI: 10.1113/jphysiol.2014.280065] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The epithalamic lateral habenula (LHb) is implicated as part of the mammalian brain's circadian system. Anatomical evidence suggests that the LHb receives extrinsic circadian timing cues from retinal ganglion cells and the master clock in the suprachiasmatic nuclei (SCN). Intriguingly, some LHb neurones contain the molecular circadian clock, but it is unclear if and how intrinsic and extrinsic circadian processes influence neuronal activity in the mouse LHb. Here, using an in vitro brain slice preparation isolating the LHb from the SCN, we show through whole-cell patch-clamp recordings that LHb neurones exhibit heterogeneity in their resting state, but the majority spontaneously fire action potentials (APs). Discharge rate of APs varied from low firing in the early day to higher firing later in the day and was absent in LHb brain slices prepared from Cry1(-/-)Cry2(-/-) mice that lack a functional molecular clock. Low amplitude circadian oscillations in the molecular circadian clock were also monitored in LHb brain slices, but were absent in Cry1(-/-)Cry2(-/-) LHb brain tissue. A putative neurochemical output signal of the SCN, prokineticin 2 (PK2), inhibited some LHb neurones by elevating the frequency of GABA release in the LHb. Using multi-electrode recordings in vivo, we found that LHb neurones sluggishly respond to retinal illumination, suggesting that they receive such information through polysynaptic processes. In summary, our results show for the first time that intrinsic circadian signals are important for regulating LHb neuronal state, while the SCN-derived signal PK2 is less influential. Moreover, we demonstrate that mouse LHb neurones have access to and can respond to visual input, but such signals are unlikely to be directly communicated to the LHb. Broadly, these findings raise the possibility that intrinsic circadian signals are likely to be influential in shaping LHb contributions to cognition and emotionality.
Collapse
Affiliation(s)
- Kanwal Sakhi
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - Sven Wegner
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - Mino D C Belle
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - Michael Howarth
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - Philippe Delagrange
- Unité de Recherches et Découvertes en Neurosciences, Institut de Recherches Servier, 78290, Croissy-sur-Seine, France
| | - Timothy M Brown
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - Hugh D Piggins
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
9
|
Schwartz TL, Goradia V. Managing insomnia: an overview of insomnia and pharmacologic treatment strategies in use and on the horizon. Drugs Context 2013; 2013:212257. [PMID: 24432044 PMCID: PMC3884958 DOI: 10.7573/dic.212257] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 08/30/2013] [Accepted: 09/06/2012] [Indexed: 11/21/2022] Open
Abstract
This review explores basic sleep physiology, the mechanism of action for each class of hypnotic agents, their clinical application based on pharmacodynamic and pharmacokinetic factors, and potential pharmacologic sleep-inducing mechanisms of future hypnotics. The paper challenges the reader to understand the neuroscientific basis of insomnia and use this knowledge to guide prescription of hypnotic agents. Currently indicated hypnotic agents are discussed with regard to their mechanism of drug action and clinical application. A broader discussion is developed throughout this paper regarding other non-indicated agents that may improve sleep and describing newer pharmacological treatments that may become available in the future for use in sleep disorders and their comorbid conditions.
Collapse
Affiliation(s)
- Thomas L Schwartz
- SUNY Upstate Medical University, Psychiatry Department, Syracuse, NY 13210, USA
| | - Viral Goradia
- SUNY Upstate Medical University, Psychiatry Department, Syracuse, NY 13210, USA
| |
Collapse
|
10
|
Prokineticin 2 is an endangering mediator of cerebral ischemic injury. Proc Natl Acad Sci U S A 2012; 109:5475-80. [PMID: 22431614 DOI: 10.1073/pnas.1113363109] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Stroke causes brain dysfunction and neuron death, and the lack of effective therapies heightens the need for new therapeutic targets. Here we identify prokineticin 2 (PK2) as a mediator for cerebral ischemic injury. PK2 is a bioactive peptide initially discovered as a regulator of gastrointestinal motility. Multiple biological roles for PK2 have been discovered, including circadian rhythms, angiogenesis, and neurogenesis. However, the role of PK2 in neuropathology is unknown. Using primary cortical cultures, we found that PK2 mRNA is up-regulated by several pathological stressors, including hypoxia, reactive oxygen species, and excitotoxic glutamate. Glutamate-induced PK2 expression is dependent on NMDA receptor activation and extracellular calcium. Enriched neuronal culture studies revealed that neurons are the principal source of glutamate-induced PK2. Using in vivo models of stroke, we found that PK2 mRNA is induced in the ischemic cortex and striatum. Central delivery of PK2 worsens infarct volume, whereas PK2 receptor antagonist decreases infarct volume and central inflammation while improving functional outcome. Direct central inhibition of PK2 using RNAi also reduces infarct volume. These findings indicate that PK2 can be activated by pathological stimuli such as hypoxia-ischemia and excitotoxic glutamate and identify PK2 as a deleterious mediator for cerebral ischemia.
Collapse
|
11
|
Ren P, Zhang H, Qiu F, Liu YQ, Gu H, O'Dowd DK, Zhou QY, Hu WP. Prokineticin 2 regulates the electrical activity of rat suprachiasmatic nuclei neurons. PLoS One 2011; 6:e20263. [PMID: 21687716 PMCID: PMC3110640 DOI: 10.1371/journal.pone.0020263] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Accepted: 04/21/2011] [Indexed: 12/11/2022] Open
Abstract
Neuropeptide signaling plays roles in coordinating cellular activities and maintaining robust oscillations within the mammalian suprachiasmatic nucleus (SCN). Prokineticin2 (PK2) is a signaling molecule from the SCN and involves in the generation of circadian locomotor activity. Prokineticin receptor 2 (PKR2), a receptor for PK2, has been shown to be expressed in the SCN. However, very little is known about the cellular action of PK2 within the SCN. In the present study, we investigated the effect of PK2 on spontaneous firing and miniature inhibitory postsynaptic currents (mIPSCs) using whole cell patch-clamp recording in the SCN slices. PK2 dose-dependently increased spontaneous firing rates in most neurons from the dorsal SCN. PK2 acted postsynaptically to reduce γ-aminobutyric acid (GABA)-ergic function within the SCN, and PK2 reduced the amplitude but not frequency of mIPSCs. Furthermore, PK2 also suppressed exogenous GABA-induced currents. And the inhibitory effect of PK2 required PKC activation in the postsynaptic cells. Our data suggest that PK2 could alter cellular activities within the SCN and may influence behavioral and physiological rhythms.
Collapse
Affiliation(s)
- Ping Ren
- Department of Pharmacology, Xianning College, Xianning, Hubei, People's Republic of China
| | - Huiping Zhang
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Fang Qiu
- Department of Pharmacology, Xianning College, Xianning, Hubei, People's Republic of China
| | - Yu-Qiang Liu
- Department of Pharmacology, Xianning College, Xianning, Hubei, People's Republic of China
| | - Huaiyu Gu
- Departments of Anatomy and Neurobiology, Developmental and Cell Biology, University of California Irvine, Irvine, California, United States of America
| | - Diane K. O'Dowd
- Departments of Anatomy and Neurobiology, Developmental and Cell Biology, University of California Irvine, Irvine, California, United States of America
| | - Qun-Yong Zhou
- Department of Pharmacology, University of California Irvine, Irvine, California, United States of America
| | - Wang-Ping Hu
- Department of Pharmacology, Xianning College, Xianning, Hubei, People's Republic of China
- * E-mail:
| |
Collapse
|
12
|
Smith PM, Ferguson AV. Circulating signals as critical regulators of autonomic state--central roles for the subfornical organ. Am J Physiol Regul Integr Comp Physiol 2010; 299:R405-15. [PMID: 20463185 DOI: 10.1152/ajpregu.00103.2010] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
To maintain homeostasis autonomic control centers in the hypothalamus and medulla must respond appropriately to both external and internal stimuli. Although protected behind the blood-brain barrier, neurons in these autonomic control centers are known to be influenced by changing levels of important signaling molecules in the systemic circulation (e.g., osmolarity, glucose concentrations, and regulatory peptides). The subfornical organ belongs to a group of specialized central nervous system structures, the circumventricular organs, which are characterized by the lack of the normal blood-brain barrier, such that circulating lipophobic substances may act on neurons within this region and via well-documented efferent neural projections to hypothalamic autonomic control centers, influence autonomic function. This review focuses on the role of the subfornical organ in sensing peripheral signals and transmitting this information to autonomic control centers in the hypothalamus.
Collapse
Affiliation(s)
- Pauline M Smith
- Dept. of Physiology, Queen's Univ., Kingston, Ontario, Canada K7L 3N6
| | | |
Collapse
|
13
|
Ingves MV, Ferguson AV. Prokineticin 2 modulates the excitability of area postrema neurons in vitro in the rat. Am J Physiol Regul Integr Comp Physiol 2010; 298:R617-26. [DOI: 10.1152/ajpregu.00620.2009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Despite recent evidence describing prokineticin 2 (PK2)-producing neurons and receptors in the dorsomedial medulla, little is known regarding the potential mechanisms by which this circadian neuropeptide acts in the medulla to influence autonomic function. Using whole cell electrophysiology, we have investigated a potential role for PK2 in the regulation of excitability in neurons of the area postrema (AP), a medullary structure known to influence autonomic processes in the central nervous system. In current-clamp recordings, focal application of 1 μM PK2 reversibly influenced the excitability of the majority of dissociated AP cells tested, producing depolarizations (38%) and hyperpolarizations (28%) in a concentration-dependent manner. Slow voltage ramps and ion-substitution experiments revealed that a PK2-induced Cl− current was responsible for membrane depolarization, whereas hyperpolarizations were the result of inhibition of a nonselective cation current. In contrast to these differential effects on membrane potential, nearly all neurons that displayed spontaneous activity responded to PK2 with a decrease in spike frequency. These observations are in accordance with voltage-clamp experiments showing that PK2 caused a leftward shift in Na+ channel activation and inactivation gating. Lastly, using post hoc single-cell RT-PCR technology, we have shown that 7 of 10 enkephalin-expressing AP neurons were depolarized by PK2 indicating that PK2 may have specific inhibitory actions on this population of neurons in the AP to reduce their sensitivity to homeostatic signals. These data suggest that the level of AP neuronal excitability may be regulated by PK2, ultimately affecting AP autonomic control.
Collapse
Affiliation(s)
- Matthew V. Ingves
- Department of Physiology, Queen's University, Kingston, Ontario, Canada
| | | |
Collapse
|
14
|
Smith PM, Chambers AP, Price CJ, Ho W, Hopf C, Sharkey KA, Ferguson AV. The subfornical organ: a central nervous system site for actions of circulating leptin. Am J Physiol Regul Integr Comp Physiol 2008; 296:R512-20. [PMID: 19020290 DOI: 10.1152/ajpregu.90858.2008] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Adipose tissue plays a critical role in energy homeostasis, secreting adipokines that control feeding, thermogenesis, and neuroendocrine function. Leptin is the prototypic adipokine that acts centrally to signal long-term energy balance. While hypothalamic and brain stem nuclei are well-established sites of action of leptin, we tested the hypothesis that leptin signaling occurs in the subfornical organ (SFO). The SFO is a circumventricular organ (CVO) that lacks the normal blood-brain barrier, is an important site in central autonomic regulation, and has been suggested to have a role in modulating peripheral signals indicating energy status. We report here the presence of mRNA for the signaling form of the leptin receptor in SFO and leptin receptor localization by immunohistochemistry within this CVO. Central administration of leptin resulted in phosphorylation of STAT3 in neurons of SFO. Whole cell current-clamp recordings from dissociated SFO neurons demonstrated that leptin (10 nM) influenced the excitability of 64% (46/72) of SFO neurons. Leptin was found to depolarize the majority of responsive neurons with a mean change in membrane potential of 7.3 +/- 0.6 mV (39% of all SFO neurons), while the remaining cells that responded to leptin hyperpolarized (-6.9 +/- 0.7 mV, 25% of all SFO neurons). Similar depolarizing and hyperpolarizing effects of leptin were observed in recordings from acutely prepared SFO slice preparations. Leptin was found to influence the same population of SFO neurons influenced by amylin as three of four cells tested for the effects of bath application of both amylin and leptin depolarized to both peptides. These observations identify the SFO as a possible central nervous system location, with direct access to the peripheral circulation, at which leptin may act to influence hypothalamic control of energy homeostasis.
Collapse
Affiliation(s)
- P M Smith
- Dept of Physiology, Queen's Univ., Kingston, Ontario, Canada K7L 3N6
| | | | | | | | | | | | | |
Collapse
|
15
|
Fry M, Cottrell GT, Ferguson AV. Prokineticin 2 influences subfornical organ neurons through regulation of MAP kinase and the modulation of sodium channels. Am J Physiol Regul Integr Comp Physiol 2008; 295:R848-56. [DOI: 10.1152/ajpregu.00779.2007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Prokineticin 2 (PK2) is a neuropeptide that acts as a signaling molecule regulating circadian rhythms in mammals. We have previously reported PK2 actions on subfornical organ (SFO) neurons, identifying this circumventricular organ as a target at which PK2 acts to influence autonomic control (Cottrell GT, and Ferguson AV. J. Neurosci. 24: 2375–2379, 2004). In this study, we have examined the cellular mechanisms by which PK2 increases the excitability of SFO neurons. Whole cell patch recordings from dissociated rat SFO neurons demonstrated that the mitogen-activated protein (MAP) kinase inhibitor PD-98059 prevented PK2-induced depolarization and decreases in delayed rectifier K+ current. PK2 also increased intracellular Ca2+ concentration ([Ca2+]i) in 39% of dissociated SFO neurons (mean increase = 20.8 ± 5.5%), effects that were maintained in the presence of thapsigargin but abolished by both nifedipine, or the absence of extracellular Ca2+, suggesting that PK2-induced [Ca2+]i transients resulted from Ca2+ entry through voltage-gated Ca2+ channels. Voltage-clamp recordings showed that PK2 was without effects on Ca2+ currents evoked by voltage ramps, suggesting that PK2-induced Ca2+ influx was secondary to PK2-induced increases in action potential frequency, an hypothesis supported by data showing that tetrodotoxin abolished effects of PK2 on [Ca2+]i. These observations suggested PK2 modulation of voltage-gated Na+ currents, a possibility confirmed by voltage-clamp experiments showing that PK2 increased the amplitude of both transient and persistent Na+ currents in 29% of SFO neurons (by 34 and 38%, respectively). These data indicate that PK2 influences SFO neurons through the activation of a MAP kinase cascade, which, in turn, modulates Na+ and K+ conductances.
Collapse
|
16
|
Abstract
Secreted peptides have been implicated in diverse physiological functions. Prokineticins are a pair of regulatory peptides that signal through two highly homologous G protein-coupled receptors. Prokineticins possess a unique structural motif of five disulfide bonds and conserved N-terminal stretches. Diverse biological functions, ranging from development to adult physiology, have been attributed to prokineticins. Herein we provide an overview of current knowledge of this interesting pair of regulatory peptides.
Collapse
Affiliation(s)
- Q-Y Zhou
- Department of Pharmacology, University of California, Irvine, CA 92697, USA.
| | | |
Collapse
|
17
|
Negri L, Lattanzi R, Giannini E, Melchiorri P. Bv8/Prokineticin proteins and their receptors. Life Sci 2007; 81:1103-16. [PMID: 17881008 DOI: 10.1016/j.lfs.2007.08.011] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2007] [Revised: 08/04/2007] [Accepted: 08/08/2007] [Indexed: 11/23/2022]
Abstract
The Bv8/Prokineticins (PKs) are a new family of peptides identified in frog, fish, reptiles and mammals that signal through two highly homologous G-protein coupled receptors, PKR1 and PKR2. Bv8/PK proteins possess a unique structural motif comprising five disulfide bonds and a completely conserved N-terminal hexapeptide sequence that is essential for the peptide's biological activities. Over the past few years, several biological functions of Bv8/PK proteins have been elucidated. This review considers all the published data on the action and physiological role of this new biological system implicated in angiogenesis and neurogenesis, in reproduction and cancer and in regulating physiological functions that underly circadian rhythms, such as the sleep/wake cycle, hormone secretion and ingestive behaviors. The high expression level of human Bv8/PK2 in bone marrow, lymphoid organs and leukocytes suggested an involvement of these peptides in hematopoiesis and in inflammatory and immunomodulatory processes. Our review highlights the role of the Bv8/PK and their receptor system in setting the pain threshold under normal and pathological conditions.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Circadian Rhythm/physiology
- Gastrointestinal Hormones/chemistry
- Gastrointestinal Hormones/genetics
- Gastrointestinal Hormones/metabolism
- Gastrointestinal Motility/physiology
- Humans
- Inflammation
- Mice
- Molecular Sequence Data
- Molecular Structure
- Neovascularization, Pathologic/metabolism
- Neovascularization, Physiologic
- Neuropeptides/chemistry
- Neuropeptides/genetics
- Neuropeptides/metabolism
- Pain Threshold/physiology
- Rabbits
- Rats
- Receptors, G-Protein-Coupled/chemistry
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Peptide/chemistry
- Receptors, Peptide/genetics
- Receptors, Peptide/metabolism
- Signal Transduction
- Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/chemistry
- Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/genetics
- Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/metabolism
Collapse
Affiliation(s)
- Lucia Negri
- Department of Human Physiology and Pharmacology "V. Erspamer", University "La Sapienza", P.le A: Moro 5, 00185 Rome, Italy.
| | | | | | | |
Collapse
|
18
|
Guilding C, Piggins HD. Challenging the omnipotence of the suprachiasmatic timekeeper: are circadian oscillators present throughout the mammalian brain? Eur J Neurosci 2007; 25:3195-216. [PMID: 17552989 DOI: 10.1111/j.1460-9568.2007.05581.x] [Citation(s) in RCA: 237] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The suprachiasmatic nucleus of the hypothalamus (SCN) is the master circadian pacemaker or clock in the mammalian brain. Canonical theory holds that the output from this single, dominant clock is responsible for driving most daily rhythms in physiology and behaviour. However, important recent findings challenge this uniclock model and reveal clock-like activities in many neural and non-neural tissues. Thus, in addition to the SCN, a number of areas of the mammalian brain including the olfactory bulb, amygdala, lateral habenula and a variety of nuclei in the hypothalamus, express circadian rhythms in core clock gene expression, hormone output and electrical activity. This review examines the evidence for extra-SCN circadian oscillators in the mammalian brain and highlights some of the essential properties and key differences between brain oscillators. The demonstration of neural pacemakers outside the SCN has wide-ranging implications for models of the circadian system at a whole-organism level.
Collapse
Affiliation(s)
- Clare Guilding
- 3.614 Stopford Building, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | | |
Collapse
|
19
|
Yuill EA, Hoyda TD, Ferri CC, Zhou QY, Ferguson AV. Prokineticin 2 depolarizes paraventricular nucleus magnocellular and parvocellular neurons. Eur J Neurosci 2007; 25:425-34. [PMID: 17284183 PMCID: PMC2667317 DOI: 10.1111/j.1460-9568.2006.05293.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Blind whole-cell patch-clamp techniques were used to examine the effects of prokineticin 2 (PK2) on the excitability of magnocellular (MNC), parvocellular preautonomic (PA), and parvocellular neuroendocrine (NE) neurons within the hypothalamic paraventricular nucleus (PVN) of the rat. The majority of MNC neurons (76%) depolarized in response to 10 nm PK2, effects that were eliminated in the presence of tetrodotoxin (TTX). PK2 also caused an increase in excitatory postsynaptic potential (EPSP) frequency, a finding that was confirmed by voltage clamp recordings demonstrating increases in excitatory postsynaptic current (EPSC) frequency. The depolarizing effects of PK2 on MNC neurons were also abolished by kynurenic acid (KA), supporting the conclusion that the effects of PK2 are mediated by the activation of glutamate interneurons within the hypothalamic slice. PA (68%) and NE (67%) parvocellular neurons also depolarized in response to 10 nm PK2. However, in contrast to MNC neurons, these effects were maintained in TTX, indicating that PK2 directly affects PA and NE neurons. PK2-induced depolarizations observed in PA and NE neurons were found to be concentration-related and receptor mediated, as experiments performed in the presence of A1MPK1 (a PK2 receptor antagonist) abolished the effects of PK2 on these subpopulations of neurons. The depolarizing effects of PK2 on PA and NE neurons were also shown to be abolished by PD 98059 (a mitogen activated protein kinase (MAPK) inhibitor) suggesting that PK2 depolarizes PVN parvocellular neurons through a MAPK signalling mechanism. In combination, these studies have identified separate cellular mechanisms through which PK2 influences the excitability of different subpopulations of PVN neurons.
Collapse
Affiliation(s)
- Erik A Yuill
- Department of Physiology, Queen's University, Kingston, Ontario, Canada
| | | | | | | | | |
Collapse
|
20
|
Abstract
Secreted peptides play broad regulatory roles in brain function and elsewhere in the body. Prokineticins are a pair of newly identified regulatory peptides that signal through two highly homologous G protein-coupled receptors. Prokineticins possess a unique structural motif of five disulfide bonds and a completely conserved N-terminal hexapeptide sequence that is essential to biological activity. Diverse biological functions, including roles in development and cell differentiation, have been assigned to the prokineticins. A network of genes, subject to various transcriptional factors, may functionally converge on the prokineticins as regulatory targets.
Collapse
Affiliation(s)
- Qun-Yong Zhou
- Department of Pharmacology, University of California, Irvine, California 92697, USA.
| |
Collapse
|
21
|
Hu WP, Zhang C, Li JD, Luo ZD, Amadesi S, Bunnett N, Zhou QY. Impaired pain sensation in mice lacking prokineticin 2. Mol Pain 2006; 2:35. [PMID: 17107623 PMCID: PMC1660571 DOI: 10.1186/1744-8069-2-35] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2006] [Accepted: 11/15/2006] [Indexed: 01/08/2023] Open
Abstract
Prokineticins (PKs), consisting of PK1 and PK2, are a pair of newly identified regulatory peptides. Two closely related G-protein coupled receptors, PKR1 and PKR2, mediate the signaling of PKs. PKs/PKRs participate in the regulation of diverse biological processes, ranging from development to adult physiology. A number of studies have indicated the involvement of PKs/PKRs in nociception. Here we show that PK2 is a sensitizer for nociception. Intraplantar injection of recombinant PK2 resulted in a strong and localized hyperalgesia with reduced thresholds to nociceptive stimuli. PK2 mobilizes calcium in dissociated dorsal root ganglion (DRG) neurons. Mice lacking the PK2 gene displayed strong reduction in nociception induced by thermal and chemical stimuli, including capsaicin. However, PK2 mutant mice showed no difference in inflammatory response to capsaicin. As the majority of PK2-responsive DRG neurons also expressed transient receptor potential vanilloid (TRPV1) and exhibited sensitivity to capsaicin, TRPV1 is likely a significant downstream molecule of PK2 signaling. Taken together, these results reveal that PK2 sensitize nociception without affecting inflammation.
Collapse
Affiliation(s)
- Wang-Ping Hu
- Department of Pharmacology, University of California, Irvine, CA 92697, USA
| | - Chengkang Zhang
- Department of Pharmacology, University of California, Irvine, CA 92697, USA
| | - Jia-Da Li
- Department of Pharmacology, University of California, Irvine, CA 92697, USA
| | - Z David Luo
- Department of Pharmacology, University of California, Irvine, CA 92697, USA
- Department of Anesthesiology, University of California, Irvine, CA 92697, USA
| | - Silvia Amadesi
- Departments of Surgery and Physiology, University of California, San Francisco, CA 94143, USA
| | - Nigel Bunnett
- Departments of Surgery and Physiology, University of California, San Francisco, CA 94143, USA
| | - Qun-Yong Zhou
- Department of Pharmacology, University of California, Irvine, CA 92697, USA
| |
Collapse
|
22
|
CHENG MICHELLEY, LESLIE FRANCESM, ZHOU QUNYONG. Expression of prokineticins and their receptors in the adult mouse brain. J Comp Neurol 2006; 498:796-809. [PMID: 16927269 PMCID: PMC2667319 DOI: 10.1002/cne.21087] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Prokineticins are a pair of regulatory peptides that have been shown to play important roles in gastrointestinal motility, angiogenesis, circadian rhythms, and, recently, olfactory bulb neurogenesis. Prokineticins exert their functions via activation of two closely related G-protein-coupled receptors. Here we report a comprehensive mRNA distribution for both prokineticins (PK1 and PK2) and their receptors (PKR1 and PKR2) in the adult mouse brain with the use of in situ hybridization. PK2 mRNA is expressed in discrete regions of the brain, including suprachiasmatic nucleus, islands of Calleja and medial preoptic area, olfactory bulb, nucleus accumbens shell, hypothalamic arcuate nucleus, and amygdala. PK1 mRNA is expressed exclusively in the brainstem, with high abundance in the nucleus tractus solitarius. PKR2 mRNA is detected throughout the brain, with prominent expression in olfactory regions, cortex, thalamus and hypothalamus, septum and hippocampus, habenula, amygdala, nucleus tractus solitarius, and circumventricular organs such as subfornical organ, median eminence, and area postrema. PKR2 mRNA is also detected in mammillary nuclei, periaqueductal gray, and dorsal raphe. In contrast, PKR1 mRNA is found in fewer brain regions, with moderate expression in the olfactory regions, dentate gyrus, zona incerta, and dorsal motor vagal nucleus. Both PKR1 and PKR2 are also detected in olfactory ventricle and subventricular zone of the lateral ventricle, both of which are rich sources of neuronal precursors. These extensive expression patterns suggest that prokineticins may have a broad array of functions in the central nervous system, including circadian rhythm, neurogenesis, ingestive behavior, reproduction, and autonomic function.
Collapse
Affiliation(s)
- MICHELLE Y. CHENG
- Department of Pharmacology, University of California, Irvine, California 92697-4625
| | - FRANCES M. LESLIE
- Department of Pharmacology, University of California, Irvine, California 92697-4625
- Department of Anatomy and Neurobiology, University of California, Irvine, California 92697-4625
| | - QUN-YONG ZHOU
- Department of Pharmacology, University of California, Irvine, California 92697-4625
- Correspondence to: Qun-Yong Zhou, 363D Med Surge II, Irvine, CA 92697-4625. E-mail:
| |
Collapse
|
23
|
Masumoto KH, Nagano M, Takashima N, Hayasaka N, Hiyama H, Matsumoto SI, Inouye SIT, Shigeyoshi Y. Distinct localization of prokineticin 2 and prokineticin receptor 2 mRNAs in the rat suprachiasmatic nucleus. Eur J Neurosci 2006; 23:2959-70. [PMID: 16819985 DOI: 10.1111/j.1460-9568.2006.04834.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The suprachiasmatic nucleus (SCN) is the master circadian clock that regulates physiological and behavioral circadian rhythms in mammals. Prokineticin 2 (PK2) is highly expressed in the SCN, and its involvement in the generation of circadian locomotor activity has been reported previously. In the present study, using in situ hybridization methods, we investigated the localization of PK2 and prokineticin receptor 2 (PKR2), a specific receptor for PK2, in the rat SCN. In steady light : dark (L : D = 12 : 12 h) and constant dark conditions, rPK2 mRNA displayed a robust circadian oscillation with a peak occurring during the day. Moreover, during peak expression, the rPK2 mRNA-positive neurons were scattered in both the dorsomedial and ventrolateral SCN, which are two functionally and morphologically distinct subregions. Furthermore, double-labeling in situ hybridization experiments revealed that greater than 50% of the rPK2 mRNA-containing neurons co-expressed either vasoactive intestinal peptide (VIP), gastrin-releasing peptide (GRP) or arginine vasopressin (AVP) in the SCN. In contrast, the rPKR2 mRNA levels did not show significant diurnal alterations. rPKR2 mRNA-containing neurons were also clustered in the dorsolateral part of the SCN, which shows negligible labeling of either rAVP, rVIP, rGRP or rPK2 transcripts. In addition, this region exhibited a delayed cycling of the rPer1 gene. These results suggest an intrinsic PK2 neurotransmission and functionally distinct roles for PKR2-expressing neurons in the SCN.
Collapse
Affiliation(s)
- Koh-hei Masumoto
- Department of Physics, Informatics and Biology, Yamaguchi University, Yoshida, Yamaguchi 753-8512, Japan
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Koyama Y, Kiyo-oka M, Osakada M, Horiguchi N, Shintani N, Ago Y, Kakuda M, Baba A, Matsuda T. Expression of prokineticin receptors in mouse cultured astrocytes and involvement in cell proliferation. Brain Res 2006; 1112:65-9. [PMID: 16901473 DOI: 10.1016/j.brainres.2006.07.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2006] [Revised: 06/30/2006] [Accepted: 07/04/2006] [Indexed: 11/30/2022]
Abstract
Effects of prokineticins (PKs), a novel family of bioactive peptides with a mitogenic action to endothelial cells of the endocrine gland and testis, on astrocytic functions were examined. Mouse cultured astrocytes expressed PK-R1 type PK receptors, while there was little expression of the PK-R2 type. PKs caused increases in astrocytic cytosolic Ca2+ levels and BrdU incorporation. Increases in Ca2+ levels by PK-2 were diminished by U73122 (a phospholipase C inhibitor). PK-induced BrdU incorporation was inhibited by U73122, GF109203 (a protein kinase C inhibitor) and PD98059 (a MEK/ERK inhibitor). These results indicate that PK receptors are expressed in astrocytes and regulate astrocytic proliferation.
Collapse
Affiliation(s)
- Yutaka Koyama
- Laboratory of Medicinal Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Yamada-Oka 1-6 Suita, Osaka 565-0871, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Pulman KJ, Fry WM, Cottrell GT, Ferguson AV. The subfornical organ: a central target for circulating feeding signals. J Neurosci 2006; 26:2022-30. [PMID: 16481435 PMCID: PMC6674925 DOI: 10.1523/jneurosci.3218-05.2006] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2005] [Revised: 01/05/2005] [Accepted: 01/06/2006] [Indexed: 11/21/2022] Open
Abstract
The mechanisms through which circulating ghrelin relays hunger signals to the CNS are not yet fully understood. In this study, we have examined the potential role of the subfornical organ (SFO), a circumventricular structure that lacks the normal blood-brain barrier, as a CNS site in which ghrelin acts to influence the hypothalamic centers controlling food intake. We report that ghrelin increased intracellular calcium concentrations in 28% (12 of 43) of dissociated SFO neurons and that the SFO expresses mRNA for the growth hormone secretagogue receptor. Whole-cell patch recordings from SFO neurons demonstrated that in 29% (9 of 31) of neurons tested ghrelin induced a mean depolarization of 7.4 +/- 0.69 mV, accompanied by an increase in action potential frequency. Voltage-clamp recordings revealed that ghrelin activates a putative nonselective cationic conductance. Previous reports that the satiety signal amylin exerts similar excitatory effects on SFO neurons led us to examine whether these two peptides influence different subpopulations of SFO neurons. Concentration-dependent depolarizing effects of amylin were observed in 59% (28 of 47) of SFO neurons (mean depolarization, 8.32 +/- 0.60 mV). In contrast to ghrelin, voltage-clamp recordings suggest that amylin influences a voltage-dependent current activated at depolarized potentials. We tested single SFO neurons with both peptides and identified cells responsive only to ghrelin (n = 9) and only to amylin (n = 7) but no cells that responded to both peptides. These data support a role for the SFO as a center at which ghrelin and amylin may influence separate subpopulations of neurons to influence the hypothalamic regulation of feeding.
Collapse
|
26
|
Yi CX, van der Vliet J, Dai J, Yin G, Ru L, Buijs RM. Ventromedial arcuate nucleus communicates peripheral metabolic information to the suprachiasmatic nucleus. Endocrinology 2006; 147:283-94. [PMID: 16195398 DOI: 10.1210/en.2005-1051] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The arcuate nucleus (ARC) is crucial for the maintenance of energy homeostasis as an integrator of long- and short-term hunger and satiety signals. The expression of receptors for metabolic hormones, such as insulin, leptin, and ghrelin, allows ARC to sense information from the periphery and signal it to the central nervous system. The ventromedial ARC (vmARC) mainly comprises orexigenic neuropeptide agouti-related peptide and neuropeptide Y neurons, which are sensitive to circulating signals. To investigate neural connections of vmARC within the central nervous system, we injected the neuronal tracer cholera toxin B into vmARC. Due to variation of injection sites, tracer was also injected into the subependymal layer of the median eminence (seME), which showed similar projection patterns as the vmARC. We propose that the vmARC forms a complex with the seME, their reciprocal connections with viscerosensory areas in brain stem, and other circumventricular organs, suggesting the exchange of metabolic and circulating information. For the first time, the vmARC-seME was shown to have reciprocal interaction with the suprachiasmatic nucleus (SCN). Activation of vmARC neurons by systemic administration of the ghrelin mimetic GH-releasing peptide-6 combined with SCN tracing showed vmARC neurons to transmit feeding related signals to the SCN. The functionality of this pathway was demonstrated by systemic injection of GH-releasing peptide-6, which induced Fos in the vmARC and resulted in a reduction of about 40% of early daytime Fos immunoreactivity in the SCN. This observation suggests an anatomical and functional pathway for peripheral hormonal feedback to the hypothalamus, which may serve to modulate the activity of the SCN.
Collapse
Affiliation(s)
- Chun-Xia Yi
- Netherlands Institute for Brain Research, Meibergdreef 33, Amsterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
27
|
Buijs RM, Scheer FA, Kreier F, Yi C, Bos N, Goncharuk VD, Kalsbeek A. Organization of circadian functions: interaction with the body. PROGRESS IN BRAIN RESEARCH 2006; 153:341-60. [PMID: 16876585 DOI: 10.1016/s0079-6123(06)53020-1] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The hypothalamus integrates information from the brain and the body; this activity is essential for survival of the individual (adaptation to the environment) and the species (reproduction). As a result, countless functions are regulated by neuroendocrine and autonomic hypothalamic processes in concert with the appropriate behaviour that is mediated by neuronal influences on other brain areas. In the current chapter attention will be focussed on fundamental hypothalamic systems that control metabolism, circulation and the immune system. Herein a system is defined as a physiological and anatomical functional unit, responsible for the organisation of one of these functions. Interestingly probably because these systems are essential for survival, their function is highly dependent on each other's performance and often shares same hypothalamic structures. The functioning of these systems is strongly influenced by (environmental) factors such as the time of the day, stress and sensory autonomic feedback and by circulating hormones. In order to get insight in the mechanisms of hypothalamic integration we have focussed on the influence of the biological clock; the suprachiasmatic nucleus (SCN) on processes that are organized by and in the hypothalamus. The SCN imposes its rhythm onto the body via three different routes of communication: 1.Via the secretion of hormones; 2. via the parasympathetic and 3.via the sympathetic autonomous nervous system. The SCN uses separate connections via either the sympathetic or via the parasympathetic system not only to prepare the body for the coming change in activity cycle but also to prepare the body and its organs for the hormones that are associated with such change. Up till now relatively little attention has been given to the question how peripheral information might be transmitted back to the SCN. Apart from light and melatonin little is known about other systems from the periphery that may provide information to the SCN. In this chapter attention will be paid to e.g. the role of the circumventricular organs in passing info to the SCN. Herein especially the role of the arcuate nucleus (ARC) will be highlighted. The ARC is crucial in the maintenance of energy homeostasis as an integrator of long- and short-term hunger and satiety signals. Receptors for metabolic hormones like insulin, leptin and ghrelin allow the ARC to sense information from the periphery and signal it to the central nervous system. Neuroanatomical tracing studies using injections of a retrograde and anterograde tracer into the ARC and SCN showed a reciprocal connection between the ARC and the SCN which is used to transmit feeding related signals to the SCN. The implications of multiple inputs and outputs of the SCN to the body will be discussed in relation with metabolic functions.
Collapse
Affiliation(s)
- Ruud M Buijs
- Unviversidad Veracruzana, Inst. Sciences de Salud, Xalapa, Mexico.
| | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Circadian timing from the suprachiasmatic nucleus (SCN) is a critical component of sleep regulation. Animal lesion and genetic studies have indicated an essential interaction between the circadian signals and the homeostatic processes that regulate sleep. Here we summarize the biological functions of prokineticins, a pair of newly discovered regulatory proteins, with focus on the circadian function of prokineticin 2 (PK2) and its potential role in sleep-wake regulation. PK2 has been shown as a candidate SCN output molecule that regulates circadian locomotor behavior. The PK2 molecular rhythm in the SCN is predominantly controlled by the circadian transcriptional/translational loops, but also regulated directly by light. The receptor for PK2 is expressed in the primary SCN output targets that regulate circadian behavior including sleep-wake. The depolarizing effect of PK2 on neurons that express PK2 receptor may represent a possible mechanism for the regulatory role of PK2 in circadian rhythms.
Collapse
Affiliation(s)
- Qun-Yong Zhou
- Department of Pharmacology, University of California, Irvine, CA 92697-4625, USA.
| | | |
Collapse
|
29
|
Kisliouk T, Podlovni H, Spanel-Borowski K, Ovadia O, Zhou QY, Meidan R. Prokineticins (endocrine gland-derived vascular endothelial growth factor and BV8) in the bovine ovary: expression and role as mitogens and survival factors for corpus luteum-derived endothelial cells. Endocrinology 2005; 146:3950-8. [PMID: 15932929 DOI: 10.1210/en.2005-0297] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A highly vascular endocrine gland, the corpus luteum (CL) is an excellent model for the study of angiogenic factors. Prokineticins (PK-1 and -2), also termed endocrine-gland-derived vascular endothelial growth factor (VEGF) and BV8 are newly identified proteins described as selective angiogenic mitogens. We previously identified PK binding sites, two closely homologous G protein-coupled receptors (PK-R1 and PK-R2) in human and bovine ovarian cells, but their function remained unknown. In this study we examined the presence and effects of PK in CL-derived endothelial and steroidogenic cell types (LEC and LSC, respectively). PK-1 mRNA was identified in CL and follicles by real-time PCR, using primers specific for the bovine PK-1 sequence (retrieved from Bos taurus whole genome shotgun database). PK were potent angiogenic mitogens for LEC; they enhanced cell proliferation, elevated [3H]thymidine incorporation, MAPK activation, and c-jun/fos mRNA expression. The effects of PK proteins on cell survival were examined by nuclear morphology (4',6-diamidino-2-phenylindole dihydrochloride staining), measurement of DNA fragmentation (terminal dUTP nucleotide end labeling assay), and caspase-3 cleavage. Results obtained by these techniques demonstrated that PK protected LEC from serum starvation-induced apoptosis. Stress conditions such as serum withdrawal, TNF-alpha, and hypoxia markedly increased PK-R2 expression, whereas mRNA levels of PK-R1 remained unchanged. These suggest that the antiapoptotic effect of PK-1 on LEC may be mediated via PK-R2. PK-1 increased VEGF mRNA expression by LSC, implying that it could also indirectly, via VEGF, affect luteal angiogenesis. Together, these findings suggest an important role for PK-1 in luteal function by acting as a mitogen and survival factor in LEC.
Collapse
Affiliation(s)
- Tatiana Kisliouk
- Department of Animal Sciences, Faculty of Agricultural, Food and Environmental Quality Sciences, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | | | | | | | | | | |
Collapse
|
30
|
Sher D, Knebel A, Bsor T, Nesher N, Tal T, Morgenstern D, Cohen E, Fishman Y, Zlotkin E. Toxic polypeptides of the hydra—a bioinformatic approach to cnidarian allomones. Toxicon 2005; 45:865-79. [PMID: 15904682 DOI: 10.1016/j.toxicon.2005.02.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2005] [Revised: 02/14/2005] [Accepted: 02/15/2005] [Indexed: 02/02/2023]
Abstract
Cnidarians such as hydrae and sea anemones are sessile, predatory, soft bodied animals which depend on offensive and defensive allomones for prey capture and survival. These allomones are distributed throughout the entire organism both in specialized stinging cells (nematocytes) and in the body tissues. The cnidarian allomonal system is composed of neurotoxins, cytolysins and toxic phospholipapses. The present bioinformatic survey was motivated by the fact that while hydrae are the most studied model cnidarian, little is known about their allomones. A large-scale EST database from Hydra magnipapillata was searched for orthologs of known cnidarian allomones, as well as for allomones found in other venomous organisms. We show that the hydrae express orthologs of cnidarian phospholipase A2 toxins and cytolysins belonging to the actinoporin family, but could not find orthologs of the 'classic' short chain neurotoxins affecting sodium and potassium conductance. Hydrae also express proteins similar to elapid-like phospholipases, CRISP proteins, Prokineticin-like polypeptides and toxic deoxyribonucleases. Our results illustrate a high level of complexity in the hydra allomonal system, suggest that several toxins represent a basal component of all cnidarian allomones, and raise the intriguing possibility that similar proteins may fulfill both endogenous and allomonal roles in cnidaria.
Collapse
Affiliation(s)
- Daniel Sher
- Department of Cell and Animal Biology, Silberman Institute of Life Sciences, Hebrew University, Jerusalem, Israel.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Regulation of prokineticin 2 expression by light and the circadian clock. BMC Neurosci 2005; 6:17. [PMID: 15762991 PMCID: PMC555564 DOI: 10.1186/1471-2202-6-17] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2004] [Accepted: 03/11/2005] [Indexed: 11/10/2022] Open
Abstract
Background The suprachiasmatic nucleus (SCN) contains the master circadian clock that regulates daily rhythms of many physiological and behavioural processes in mammals. Previously we have shown that prokineticin 2 (PK2) is a clock-controlled gene that may function as a critical SCN output molecule responsible for circadian locomotor rhythms. As light is the principal zeitgeber that entrains the circadian oscillator, and PK2 expression is responsive to nocturnal light pulses, we further investigated the effects of light on the molecular rhythm of PK2 in the SCN. In particular, we examined how PK2 responds to shifts of light/dark cycles and changes in photoperiod. We also investigated which photoreceptors are responsible for the light-induced PK2 expression in the SCN. To determine whether light requires an intact functional circadian pacemaker to regulate PK2, we examined PK2 expression in cryptochrome1,2-deficient (Cry1-/-Cry2-/-) mice that lack functional circadian clock under normal light/dark cycles and constant darkness. Results Upon abrupt shifts of the light/dark cycle, PK2 expression exhibits transients in response to phase advances but rapidly entrains to phase delays. Photoperiod studies indicate that PK2 responds differentially to changes in light period. Although the phase of PK2 expression expands as the light period increases, decreasing light period does not further condense the phase of PK2 expression. Genetic knockout studies revealed that functional melanopsin and rod-cone photoreceptive systems are required for the light-inducibility of PK2. In Cry1-/-Cry2-/- mice that lack a functional circadian clock, a low amplitude PK2 rhythm is detected under light/dark conditions, but not in constant darkness. This suggests that light can directly regulate PK2 expression in the SCN. Conclusion These data demonstrate that the molecular rhythm of PK2 in the SCN is regulated by both the circadian clock and light. PK2 is predominantly controlled by the endogenous circadian clock, while light plays a modulatory role. The Cry1-/-Cry2-/- mice studies reveal a light-driven PK2 rhythm, indicating that light can induce PK2 expression independent of the circadian oscillator. The light inducibility of PK2 suggests that in addition to its role in clock-driven rhythms of locomotor behaviour, PK2 may also participate in the photic entrainment of circadian locomotor rhythms.
Collapse
|