1
|
Cooper AH, Nie AA, Hedden NS, Herzog H, Taylor BK. De novo expression of neuropeptide Y in sensory neurons does not contribute to peripheral neuropathic pain. THE JOURNAL OF PAIN 2025; 30:105385. [PMID: 40174733 DOI: 10.1016/j.jpain.2025.105385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/04/2025]
Abstract
Nerve damage induces a robust de novo expression of the pain-modulatory peptide neuropeptide Y (NPY) in large-diameter primary afferent neurons that innervate the dorsal horn of the spinal cord and the dorsal column nuclei. To determine whether this functions to modulate peripheral neuropathic pain in male and female mice, we selectively deleted the Npy gene in neurons of the dorsal root ganglion (DRG), without disruption of its expression in brain or dorsal horn neurons. We then subjected sensory neuron-specific NPY deletion mutant mice (Pirt-NPY) and their wild-type controls to either sham surgery, spared sural nerve injury (SNI) or spared tibial nerve injury (tSNI). Conditional Npy deletion did not change the severity or duration of static mechanical, dynamic mechanical, or cold allodynia in SNI or tSNI models, nor ongoing neuropathic pain as assessed with conditioned place preference to gabapentin. When injected after the resolution of tSNI-induced mechanical hypersensitivity (a latent pain sensitization model of chronic neuropathic pain), the NPY Y1 receptor-specific antagonist BIBO3304 equally reinstated mechanical hypersensitivity in Pirt-NPY mice and their wildtype controls. We conclude that nerve injury-induced upregulation of NPY in sensory neurons does not cause mechanical or cold hypersensitivity or ongoing pain, and that tonic inhibitory control of neuropathic pain by NPY in the spinal cord is mediated by release from dorsal horn interneurons rather than sensory neurons. PERSPECTIVE: This article answers the long-standing question as to whether nerve injury-induced upregulation of NPY in primary afferent neurons modulates neuropathic pain. We report that sensory neuron-specific NPY knockout did not change pain-like behaviors. CNS interneurons rather than sensory neurons likely mediate the well-documented phenomenon of spinal NPY analgesia.
Collapse
Affiliation(s)
- A H Cooper
- Pittsburgh Center for Pain Research, Pittsburgh Project to end Opioid Misuse, Department of Anesthesiology & Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - A A Nie
- Pittsburgh Center for Pain Research, Pittsburgh Project to end Opioid Misuse, Department of Anesthesiology & Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - N S Hedden
- Pittsburgh Center for Pain Research, Pittsburgh Project to end Opioid Misuse, Department of Anesthesiology & Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - H Herzog
- St Vincent's Centre for Applied Medical Research (AMR), Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, The University of New South Wales, Sydney, Australia
| | - B K Taylor
- Pittsburgh Center for Pain Research, Pittsburgh Project to end Opioid Misuse, Department of Anesthesiology & Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine.
| |
Collapse
|
2
|
Basu P, Taylor BK. Neuropeptide Y Y2 receptors in acute and chronic pain and itch. Neuropeptides 2024; 108:102478. [PMID: 39461244 DOI: 10.1016/j.npep.2024.102478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/29/2024]
Abstract
Pain and itch are regulated by a diverse array of neuropeptides and their receptors in superficial laminae of the spinal cord dorsal horn (DH). Neuropeptide Y (NPY) is normally expressed on DH neurons but not sensory neurons. By contrast, the Npy2r receptor (Y2) is expressed on the central and peripheral terminals of sensory neurons but not on DH neurons. Neurophysiological slice recordings indicate that Y2-selective agonists inhibits spinal neurotransmitter release from sensory neurons. However, behavioral pharmacology studies indicate that Y2 agonists exert minimal changes in nociception, even after injury. Additional discrepancies in the behavioral actions of the Y2-antagonist BIIE0246 - reports of either pronociception or antinociception - have now been resolved. In the normal state, spinally-directed (intrathecal) administration of BIIE0246 elicits ongoing nociception, hypersensitivity to sensory stimulation, and aversion. Conversely, in the setting of nerve injury and inflammation, intrathecal BIIE024 reduced not only mechanical and thermal hypersensitivity, but also a measure of the affective dimension of pain (conditioned place preference). When administered in chronic pain models of latent sensitization, BIIE0246 produced a profound reinstatement of pain-like behaviors. We propose that tissue or nerve injury induces a G protein switch in the action of NPY-Y2 signaling from antinociception in the naïve state to the inhibition of mechanical and heat hyperalgesia in the injured state, and then a switch back to antinociception to keep LS in a state of remission. This model clarifies the pharmacotherapeutic potential of Y2 research, pointing to the development of a new non-opioid pharmacotherapy for chronic pain using Y2 antagonists in patients who do not develop LS.
Collapse
Affiliation(s)
- Paramita Basu
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, Pittsburgh Project to end Opioid Misuse, United States of America
| | - Bradley K Taylor
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, Pittsburgh Project to end Opioid Misuse, United States of America; Department of Pharmacology and Chemical Biology, United States of America; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States of America.
| |
Collapse
|
3
|
Basu P, Maddula A, Nelson TS, Prasoon P, Winter MK, Herzog H, McCarson KE, Taylor BK. Neuropeptide Y Y2 Receptors in Sensory Neurons Tonically Suppress Nociception and Itch but Facilitate Postsurgical and Neuropathic Pain Hypersensitivity. Anesthesiology 2024; 141:946-968. [PMID: 39121458 PMCID: PMC11461131 DOI: 10.1097/aln.0000000000005184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2024]
Abstract
BACKGROUND Neuropeptide Y (NPY) Y2 receptor (Y2) antagonist BIIE0246 can both inhibit and facilitate nociception. The authors hypothesized that Y2 function depends on inflammation or nerve injury status. METHODS The authors implemented a battery of behavioral tests in mice of both sexes that received (1) no injury; (2) an incision model of postoperative pain; (3) a spared nerve injury model of neuropathic pain; and (4) a latent sensitization model of chronic postsurgical pain. In addition to Y2 gene expression assays, spinal Y2 G-protein coupling was studied with guanosine-5'-O-(3-[35S]thio)triphosphate ([35S]GTPγS) binding assays. RESULTS The authors report that intrathecal BIIE0246 increased mechanical and cold hypersensitivity, produced behavioral signs of spontaneous nociception and itch, and produced conditioned place aversion and preference in normal, uninjured mice. BIIE0246 did not change heat hypersensitivity or motor coordination. Conditional (sensory neuron-specific) Y2 deletion prevented BIIE0246-induced mechanical and cold hypersensitivity, nocifensive behaviors, and aversion. Both conditional deletion and pharmacologic blockade of Y2 reduced mechanical and thermal hypersensitivity after incision or nerve injury. SNI did not change the sensitivity of Y2 G-protein coupling with the Y2 agonist peptide YY (3-36) (PYY3-36), but increased the population of Y2 that effectively coupled G-proteins. Intrathecal PYY3-36 failed to reduce spared nerve injury- or incision-induced hypersensitivity in C57BL/6N mice. Incision did not change Npy2r gene expression in dorsal root ganglion. CONCLUSIONS The authors conclude that Y2 at central terminals of primary afferent neurons provides tonic inhibition of mechanical and cold nociception and itch. This switches to the promotion of mechanical and thermal hyperalgesia in models of acute and chronic postsurgical and neuropathic pain, perhaps due to an increase in the population of Y2 that effectively couples to G-proteins. These results support the development of Y2 antagonists for the treatment of chronic postsurgical and neuropathic pain. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Paramita Basu
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, and Pittsburgh Project to end Opioid Misuse, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Akshitha Maddula
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, and Pittsburgh Project to end Opioid Misuse, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Tyler S. Nelson
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, and Pittsburgh Project to end Opioid Misuse, School of Medicine, University of Pittsburgh, Pittsburgh, PA
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA
- Department of Molecular Pathobiology, NYU Pain Research Center, College of Dentistry, New York University, New York, NY 10010
| | - Pranav Prasoon
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, and Pittsburgh Project to end Opioid Misuse, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Michelle K. Winter
- Kansas Intellectual and Developmental Disabilities Research Center and Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160
| | - Herbert Herzog
- Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Kenneth E. McCarson
- Kansas Intellectual and Developmental Disabilities Research Center and Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160
| | - Bradley K. Taylor
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, and Pittsburgh Project to end Opioid Misuse, School of Medicine, University of Pittsburgh, Pittsburgh, PA
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
4
|
Chen L, Hu Y, Wang S, Cao K, Mai W, Sha W, Ma H, Zeng LH, Xu ZZ, Gao YJ, Duan S, Wang Y, Gao Z. mTOR-neuropeptide Y signaling sensitizes nociceptors to drive neuropathic pain. JCI Insight 2022; 7:159247. [PMID: 36194480 DOI: 10.1172/jci.insight.159247] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 09/29/2022] [Indexed: 12/15/2022] Open
Abstract
Neuropathic pain is a refractory condition that involves de novo protein synthesis in the nociceptive pathway. The mTOR is a master regulator of protein translation; however, mechanisms underlying its role in neuropathic pain remain elusive. Using the spared nerve injury-induced neuropathic pain model, we found that mTOR was preferentially activated in large-diameter dorsal root ganglion (DRG) neurons and spinal microglia. However, selective ablation of mTOR in DRG neurons, rather than microglia, alleviated acute neuropathic pain in mice. We show that injury-induced mTOR activation promoted the transcriptional induction of neuropeptide Y (Npy), likely via signal transducer and activator of transcription 3 phosphorylation. NPY further acted primarily on Y2 receptors (Y2R) to enhance neuronal excitability. Peripheral replenishment of NPY reversed pain alleviation upon mTOR removal, whereas Y2R antagonists prevented pain restoration. Our findings reveal an unexpected link between mTOR and NPY/Y2R in promoting nociceptor sensitization and neuropathic pain.
Collapse
Affiliation(s)
- Lunhao Chen
- Spine Lab, Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yaling Hu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Siyuan Wang
- Spine Lab, Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kelei Cao
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Weihao Mai
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
| | - Weilin Sha
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong, China
| | - Huan Ma
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Ling-Hui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Zhen-Zhong Xu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Yong-Jing Gao
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Nantong, China
| | - Shumin Duan
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Yue Wang
- Spine Lab, Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihua Gao
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| |
Collapse
|
5
|
Cutaneous innervation in impaired diabetic wound healing. Transl Res 2021; 236:87-108. [PMID: 34029747 PMCID: PMC8380642 DOI: 10.1016/j.trsl.2021.05.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/09/2021] [Accepted: 05/11/2021] [Indexed: 12/11/2022]
Abstract
Type 2 diabetes is associated with several potential comorbidities, among them impaired wound healing, chronic ulcerations, and the requirement for lower extremity amputation. Disease-associated abnormal cellular responses, infection, immunological and microvascular dysfunction, and peripheral neuropathy are implicated in the pathogenesis of the wound healing impairment and the diabetic foot ulcer. The skin houses a dense network of sensory nerve afferents and nerve-derived modulators, which communicate with epidermal keratinocytes and dermal fibroblasts bidirectionally to effect normal wound healing after trauma. However, the mechanisms through which cutaneous innervation modulates wound healing are poorly understood, especially in humans. Better understanding of these mechanisms may provide the basis for targeted treatments for chronic diabetic wounds. This review provides an overview of wound healing pathophysiology with a focus on neural involvement in normal and diabetic wound healing, as well as future therapeutic perspectives to address the unmet needs of diabetic patients with chronic wounds.
Collapse
|
6
|
Chen S, Liu XY, Jiao Y, Chen ZF, Yu W. NPY2R signaling gates spontaneous and mechanical, but not thermal, pain transmission. Mol Pain 2020; 15:1744806919887830. [PMID: 31646939 PMCID: PMC6880052 DOI: 10.1177/1744806919887830] [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] [Indexed: 12/27/2022] Open
Abstract
Neuropeptide Y signaling plays an important role in inhibiting chronic
pain in the spinal cord of mice. However, little is known about the
respective roles of two major neuropeptide Y receptors, Y1R and Y2R,
in evoked and spontaneous pain behavior under normal physiological
condition. Using intrathecal administration approach, we found that
pharmacological inhibition of Y2R, unexpectedly, gave rise to
spontaneous pain behavior. In addition, Y2R antagonism also resulted
in long-lasting mechanical but not thermal hypersensitivity. By
contrast, neither overt spontaneous pain behavior nor mechanical and
thermal hypersensitivity were detected after pharmacological
inhibition of Y1R. Remarkably, the activation of Y1R produced powerful
analgesic effect: blocking both evoked and spontaneous pain behavior
resulted from Y2R antagonism. These findings highlight the pivotal
role of endogenous Y2R in gating mechanical and spontaneous pain
transmission. Importantly, our results suggest that Y1R could be a
therapeutic target that may be exploited for alleviating spontaneous
pain without affecting acute pain transmission.
Collapse
Affiliation(s)
- Sihan Chen
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China.,Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO, USA.,Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Xian-Yu Liu
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO, USA.,Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yingfu Jiao
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Zhou-Feng Chen
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO, USA.,Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA.,Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA.,Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Weifeng Yu
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| |
Collapse
|
7
|
Cellular Mechanisms for Antinociception Produced by Oxytocin and Orexins in the Rat Spinal Lamina II-Comparison with Those of Other Endogenous Pain Modulators. Pharmaceuticals (Basel) 2019; 12:ph12030136. [PMID: 31527474 PMCID: PMC6789548 DOI: 10.3390/ph12030136] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/31/2019] [Accepted: 09/12/2019] [Indexed: 01/23/2023] Open
Abstract
Much evidence indicates that hypothalamus-derived neuropeptides, oxytocin, orexins A and B, inhibit nociceptive transmission in the rat spinal dorsal horn. In order to unveil cellular mechanisms for this antinociception, the effects of the neuropeptides on synaptic transmission were examined in spinal lamina II neurons that play a crucial role in antinociception produced by various analgesics by using the whole-cell patch-clamp technique and adult rat spinal cord slices. Oxytocin had no effect on glutamatergic excitatory transmission while producing a membrane depolarization, γ-aminobutyric acid (GABA)-ergic and glycinergic spontaneous inhibitory transmission enhancement. On the other hand, orexins A and B produced a membrane depolarization and/or a presynaptic spontaneous excitatory transmission enhancement. Like oxytocin, orexin A enhanced both GABAergic and glycinergic transmission, whereas orexin B facilitated glycinergic but not GABAergic transmission. These inhibitory transmission enhancements were due to action potential production. Oxytocin, orexins A and B activities were mediated by oxytocin, orexin-1 and orexin-2 receptors, respectively. This review article will mention cellular mechanisms for antinociception produced by oxytocin, orexins A and B, and discuss similarity and difference in antinociceptive mechanisms among the hypothalamic neuropeptides and other endogenous pain modulators (opioids, nociceptin, adenosine, adenosine 5’-triphosphate (ATP), noradrenaline, serotonin, dopamine, somatostatin, cannabinoids, galanin, substance P, bradykinin, neuropeptide Y and acetylcholine) exhibiting a change in membrane potential, excitatory or inhibitory transmission in the spinal lamina II neurons.
Collapse
|
8
|
Bao L, Zhao J, Liao D, Wang G, Gregersen H. Refeeding reverses fasting-induced remodeling of afferent nerve activity in rat small intestine. Biomech Model Mechanobiol 2019; 18:1915-1926. [DOI: 10.1007/s10237-019-01185-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 06/05/2019] [Indexed: 02/08/2023]
|
9
|
Huston NJ, Brenner LA, Taylor ZC, Ritter RC. NPY2 receptor activation in the dorsal vagal complex increases food intake and attenuates CCK-induced satiation in male rats. Am J Physiol Regul Integr Comp Physiol 2019; 316:R406-R416. [PMID: 30726118 DOI: 10.1152/ajpregu.00011.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Neuropeptide Y (NPY), peptide YY (PYY), and their cognate receptors (YR) are expressed by subpopulations of central and peripheral nervous system neurons. Intracerebroventricular injections of NPY or PYY increase food intake, and intrahypothalamic NPY1 or NPY5 receptor agonist injections also increase food intake. In contrast, injection of PYY in the periphery reduces food intake, apparently by activating peripheral Y2R. The dorsal vagal complex (DVC) of the hindbrain is the site where vagal afferents relay gut satiation signals to the brain. While contributions of the DVC are increasingly investigated, a role for DVC YR in control of food intake has not been examined systematically. We used in situ hybridization to confirm expression of Y1R and Y2R, but not Y5R, in the DVC and vagal afferent neurons. We found that nanoinjections of a Y2R agonist, PYY-(3-36), into the DVC significantly increased food intake over a 4-h period in satiated male rats. PYY-(3-36)-evoked food intake was prevented by injection of a selective Y2R antagonist. Injection of a Y1R/Y5R-preferring agonist into the DVC failed to increase food intake at doses reported to increase food intake following hypothalamic injection. Finally, injection of PYY-(3-36) into the DVC prevented reduction of 30-min food intake following intraperitoneal injection of cholecystokinin (CCK). Our results indicate that activation of DVC Y2R, unlike hypothalamic or peripheral Y2R, increases food intake. Furthermore, in the context of available electrophysiological observations, our results are consistent with the hypothesis that DVC Y2R control food intake by dampening vagally mediated satiation signals in the DVC.
Collapse
Affiliation(s)
- Nathaneal J Huston
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, Washington
| | - Lynne A Brenner
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, Washington
| | - Zachary C Taylor
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, Washington
| | - Robert C Ritter
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, Washington
| |
Collapse
|
10
|
Cork SC. The role of the vagus nerve in appetite control: Implications for the pathogenesis of obesity. J Neuroendocrinol 2018; 30:e12643. [PMID: 30203877 DOI: 10.1111/jne.12643] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 09/05/2018] [Accepted: 09/05/2018] [Indexed: 12/14/2022]
Abstract
The communication between the gut and the brain is important for the control of energy homeostasis. In response to food intake, enteroendocrine cells secrete gut hormones, which ultimately suppress appetite through centrally-mediated processes. Increasing evidence implicates the vagus nerve as an important conduit in transmitting these signals from the gastrointestinal tract to the brain. Studies have demonstrated that many of the gut hormones secreted from enteroendocrine cells signal through the vagus nerve, and the sensitivity of the vagus to these signals is regulated by feeding status. Furthermore, evidence suggests that a reduction in the ability of the vagus nerve to respond to the switch between a "fasted" and "fed" state, retaining sensitivity to orexigenic signals when fed or a reduced ability to respond to satiety hormones, may contribute to obesity. This review draws together the evidence that the vagus nerve is a crucial component of appetite regulation via the gut-brain axis, with a particular emphasis on experimental techniques and future developments.
Collapse
Affiliation(s)
- Simon C Cork
- Section of Endocrinology and Investigative Medicine, Division of Endocrinology, Diabetes and Metabolism, Imperial College London, London, UK
| |
Collapse
|
11
|
Diaz-delCastillo M, Woldbye DP, Heegaard AM. Neuropeptide Y and its Involvement in Chronic Pain. Neuroscience 2018; 387:162-169. [DOI: 10.1016/j.neuroscience.2017.08.050] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 08/22/2017] [Accepted: 08/29/2017] [Indexed: 12/20/2022]
|
12
|
Page AJ, Li H. Meal-Sensing Signaling Pathways in Functional Dyspepsia. Front Syst Neurosci 2018; 12:10. [PMID: 29674959 PMCID: PMC5895752 DOI: 10.3389/fnsys.2018.00010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 03/20/2018] [Indexed: 12/12/2022] Open
Abstract
The upper gastrointestinal tract plays an important role in sensing the arrival, amount and chemical composition of a meal. Ingestion of a meal triggers a number of sensory signals in the gastrointestinal tract. These include the response to mechanical stimulation (e.g., gastric distension), from the presence of food in the gut, and the interaction of various dietary nutrients with specific "taste" receptors on specialized enteroendocrine cells in the small intestine culminating in the release of gut hormones. These signals are then transmitted to the brain where they contribute to food intake regulation by modulating appetite as well as feedback control of gastrointestinal functions (e.g., gut motility). There is evidence that the sensitivity to these food related stimuli is abnormally enhanced in functional dyspepsia leading to symptoms such nausea and bloating. In addition, these gut-brain signals can modulate the signaling pathways involved in visceral pain. This review will discuss the role of gut-brain signals in appetite regulation and the role dysregulation of this system play in functional dyspepsia.
Collapse
Affiliation(s)
- Amanda J Page
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Hui Li
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| |
Collapse
|
13
|
Kaczyńska K, Zając D, Wojciechowski P, Kogut E, Szereda-Przestaszewska M. Neuropeptides and breathing in health and disease. Pulm Pharmacol Ther 2017; 48:217-224. [PMID: 29223509 DOI: 10.1016/j.pupt.2017.12.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/20/2017] [Accepted: 12/05/2017] [Indexed: 12/18/2022]
Abstract
Regulatory neuropeptides control and regulate breathing in physiological and pathophysiological conditions. While they have been identified in the neurons of major respiratory areas, they can be active not only at the central level, but also at the periphery via chemoreceptors, vagal afferents, or locally within lungs and airways. Some neuropeptides, such as leptin or substance P, are respiratory stimulants; others, such as neurotensin, produce variable effects on respiration depending on the site of application. Some neuropeptides have been implicated in pathological states, such as obstructive sleep apnea or asthma. This article provides a concise review of the possible role and functions of several selected neuropeptides in the process of breathing in health and disease and in lung pathologies.
Collapse
Affiliation(s)
- Katarzyna Kaczyńska
- Laboratory of Respiration Physiology, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland.
| | - Dominika Zając
- Laboratory of Respiration Physiology, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| | - Piotr Wojciechowski
- Laboratory of Respiration Physiology, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| | - Ewelina Kogut
- Laboratory of Respiration Physiology, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| | | |
Collapse
|
14
|
Quallo T, Alkhatib O, Gentry C, Andersson DA, Bevan S. G protein βγ subunits inhibit TRPM3 ion channels in sensory neurons. eLife 2017; 6. [PMID: 28826490 PMCID: PMC5593501 DOI: 10.7554/elife.26138] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 07/24/2017] [Indexed: 02/07/2023] Open
Abstract
Transient receptor potential (TRP) ion channels in peripheral sensory neurons are functionally regulated by hydrolysis of the phosphoinositide PI(4,5)P2 and changes in the level of protein kinase mediated phosphorylation following activation of various G protein coupled receptors. We now show that the activity of TRPM3 expressed in mouse dorsal root ganglion (DRG) neurons is inhibited by agonists of the Gi-coupled µ opioid, GABA-B and NPY receptors. These agonist effects are mediated by direct inhibition of TRPM3 by Gβγ subunits, rather than by a canonical cAMP mediated mechanism. The activity of TRPM3 in DRG neurons is also negatively modulated by tonic, constitutive GPCR activity as TRPM3 responses can be potentiated by GPCR inverse agonists. GPCR regulation of TRPM3 is also seen in vivo where Gi/o GPCRs agonists inhibited and inverse agonists potentiated TRPM3 mediated nociceptive behavioural responses. DOI:http://dx.doi.org/10.7554/eLife.26138.001 TRPM3 belongs to a family of channel proteins that allow sodium and calcium ions to enter cells by forming pores in cell membranes. TRPM3 is found on the cell membranes of nerve cells; when ions flow into the nerves through the TRPM3 pores it triggers an electrical impulse. TRPM3 is responsible for helping us to detect heat, and mice without this protein find it difficult to sense painfully hot temperatures. Mice lacking TRPM3 also respond to other kinds of pain differently. Normally, a mouse with an injured paw becomes more sensitive to warm and hot temperatures, but this does not happen in mice that do not have TRPM3. When activated, other proteins called G-protein coupled receptors (or GPCRs for short) can make some members of this family of channel proteins more or less likely to open their pore. This in turn increases or decreases the flow of ions through the pore, respectively. Yet it was not clear if GPCRs also affect TRPM3 channels on the membranes of nerve cells. Quallo et al. have now discovered that “switching on” different GPCR proteins in sensory nerve cells from mice greatly reduces the flow of calcium ions though TRPM3 channels. The experiments made use of two pain-killing drugs, namely morphine and baclofen, and a molecule called neuropeptide Y to activate different GPCRs. GPCRs interact with a group of small proteins called G-proteins that, when activated by the receptor, split into two subunits, known as the α subunit and the βγ subunit. Once detached these subunits are free to act as messengers and interact with other proteins in the cell membrane. Quallo et al. found that TRPM3 is one of a small group of proteins that interact with the βγ subunits of the G-protein, which can explain how “switching on” GPCRs reduces the activity of TRPM3. Two independent studies by Dembla, Behrendt et al. and Badheka, Yudin et al. also report similar findings. There is currently a need to find more effective treatments for people suffering from long-term pain conditions and it has become clear that TRPM3 channels are involved in sensing both pain and temperature. These new findings show that drugs already used in the treatment of pain can dramatically change how TRPM3 works. These results might help scientists to find drugs that work in a similar way to dial down the activity of TRPM3 and to combat pain. Though first it will be important to confirm these new findings in human nerve cells. DOI:http://dx.doi.org/10.7554/eLife.26138.002
Collapse
Affiliation(s)
- Talisia Quallo
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | - Omar Alkhatib
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | - Clive Gentry
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | - David A Andersson
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | - Stuart Bevan
- Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| |
Collapse
|
15
|
Presynaptic inhibition of nociceptive neurotransmission by somatosensory neuron-secreted suppressors. SCIENCE CHINA-LIFE SCIENCES 2017. [PMID: 28624955 DOI: 10.1007/s11427-017-9061-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Noxious stimuli cause pain by activating cutaneous nociceptors. The Aδ- and C-fibers of dorsal root ganglion (DRG) neurons convey the nociceptive signals to the laminae I-II of spinal cord. In the dorsal horn of spinal cord, the excitatory afferent synaptic transmission is regulated by the inhibitory neurotransmitter γ-aminobutyric acid and modulators such as opioid peptides released from the spinal interneurons, and by serotonin, norepinepherine and dopamine from the descending inhibitory system. In contrast to the accumulated evidence for these central inhibitors and their neural circuits in the dorsal spinal cord, the knowledge about the endogenous suppressive mechanisms in nociceptive DRG neurons remains very limited. In this review, we summarize our recent findings of the presynaptic suppressive mechanisms in nociceptive neurons, the BNP/NPR-A/PKG/BKCa channel pathway, the FSTL1/α1Na+-K+ ATPase pathway and the activin C/ERK pathway. These endogenous suppressive systems in the mechanoheat nociceptors may also contribute differentially to the mechanisms of nerve injury-induced neuropathic pain or inflammation-induced pain.
Collapse
|
16
|
Visceral hyperalgesia caused by peptide YY deletion and Y2 receptor antagonism. Sci Rep 2017; 7:40968. [PMID: 28106168 PMCID: PMC5247702 DOI: 10.1038/srep40968] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 12/13/2016] [Indexed: 12/18/2022] Open
Abstract
Altered levels of colonic peptide YY (PYY) have been reported in patients suffering from functional and inflammatory bowel disorders. While the involvement of neuropeptide Y (NPY) and Y receptors in the regulation of nociception is well established, the physiological role of PYY in somatic and visceral pain is poorly understood. In this work, the role of PYY in pain sensitivity was evaluated using PYY knockout (PYY(−/−)) mice and Y2 receptor ligands. PYY(−/−) mice were more sensitive to somatic thermal pain compared to wild type (WT) mice. Visceral pain was assessed by evaluating pain-related behaviors, mouse grimace scale (MGS) and referred hyperalgesia after intrarectal administration of allyl isothiocyanate (AITC, 1 or 2%) or its vehicle, peanut oil. The pain-related behaviors induced by AITC were significantly exaggerated by PYY deletion, whereas the MGS readout and the referred hyperalgesia were not significantly affected. The Y2 receptor antagonist, BII0246, increased pain-related behaviors in response to intrarectal AITC compared to vehicle treatment while the Y2 receptor agonist, PYY(3–36), did not have a significant effect. These results indicate that endogenous PYY has a hypoalgesic effect on somatic thermal and visceral chemical pain. The effect on visceral pain seems to be mediated by peripheral Y2 receptors.
Collapse
|
17
|
Bost A, Shaib AH, Schwarz Y, Niemeyer BA, Becherer U. Large dense-core vesicle exocytosis from mouse dorsal root ganglion neurons is regulated by neuropeptide Y. Neuroscience 2017; 346:1-13. [PMID: 28089870 DOI: 10.1016/j.neuroscience.2017.01.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 01/03/2017] [Accepted: 01/04/2017] [Indexed: 12/12/2022]
Abstract
Peptidergic dorsal root ganglion (DRG) neurons transmit sensory and nociceptive information from the periphery to the central nervous system. Their synaptic activity is profoundly affected by neuromodulatory peptides stored and released from large dense-core vesicles (LDCVs). However, the mechanism of peptide secretion from DRG neurons is poorly understood. Using total internal reflection fluorescence microscopy (TIRFM), we visualized individual LDCVs loaded with fluorescent neuropeptide Y (NPY) and analyzed their stimulation-dependent release. We tested several protocols and found an overall low stimulation-secretion coupling that increased after raising intracellular Ca2+ concentration by applying a weak pre-stimulus. Interestingly, the stimulation protocol also influenced the mechanism of LDCV fusion. Depolarization of DRG neurons with a solution containing 60mM KCl triggered full fusion, kiss-and-run, and kiss-and-stay exocytosis with equal frequency. In contrast, field electrode stimulation primarily induced full fusion exocytosis. Finally, our results indicate that NPY can promote LDCV secretion. These results shed new light on the mechanism of NPY action during modulation of DRG neuron activity, an important pathway in the treatment of chronic pain.
Collapse
Affiliation(s)
- Anneka Bost
- Institute of Physiology, CIPMM, Saarland University, 66421 Homburg/Saar, Germany
| | - Ali H Shaib
- Institute of Physiology, CIPMM, Saarland University, 66421 Homburg/Saar, Germany
| | - Yvonne Schwarz
- Institute of Physiology, CIPMM, Saarland University, 66421 Homburg/Saar, Germany
| | - Barbara A Niemeyer
- Molecular Biophysics, CIPMM, Saarland University, 66421 Homburg/Saar, Germany
| | - Ute Becherer
- Institute of Physiology, CIPMM, Saarland University, 66421 Homburg/Saar, Germany.
| |
Collapse
|
18
|
Arcourt A, Gorham L, Dhandapani R, Prato V, Taberner FJ, Wende H, Gangadharan V, Birchmeier C, Heppenstall PA, Lechner SG. Touch Receptor-Derived Sensory Information Alleviates Acute Pain Signaling and Fine-Tunes Nociceptive Reflex Coordination. Neuron 2016; 93:179-193. [PMID: 27989460 DOI: 10.1016/j.neuron.2016.11.027] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 08/01/2016] [Accepted: 11/07/2016] [Indexed: 11/25/2022]
Abstract
Painful mechanical stimuli activate multiple peripheral sensory afferent subtypes simultaneously, including nociceptors and low-threshold mechanoreceptors (LTMRs). Using an optogenetic approach, we demonstrate that LTMRs do not solely serve as touch receptors but also play an important role in acute pain signaling. We show that selective activation of neuropeptide Y receptor-2-expressing (Npy2r) myelinated A-fiber nociceptors evokes abnormally exacerbated pain, which is alleviated by concurrent activation of LTMRs in a frequency-dependent manner. We further show that spatial summation of single action potentials from multiple NPY2R-positive afferents is sufficient to trigger nocifensive paw withdrawal, but additional simultaneous sensory input from LTMRs is required for normal well-coordinated execution of this reflex. Thus, our results show that combinatorial coding of noxious and tactile sensory input is required for normal acute mechanical pain signaling. Additionally, we established a causal link between precisely defined neural activity in functionally identified sensory neuron subpopulations and nocifensive behavior and pain.
Collapse
Affiliation(s)
- Alice Arcourt
- Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Louise Gorham
- Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | | | - Vincenzo Prato
- Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Francisco J Taberner
- Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Hagen Wende
- Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany; Max-Delbrueck-Center (MDC) for Molecular Medicine, Robert-Roessle-Strasse 10, 13125 Berlin, Germany
| | - Vijayan Gangadharan
- Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Carmen Birchmeier
- Max-Delbrueck-Center (MDC) for Molecular Medicine, Robert-Roessle-Strasse 10, 13125 Berlin, Germany
| | | | - Stefan G Lechner
- Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany.
| |
Collapse
|
19
|
Li CL, Li KC, Wu D, Chen Y, Luo H, Zhao JR, Wang SS, Sun MM, Lu YJ, Zhong YQ, Hu XY, Hou R, Zhou BB, Bao L, Xiao HS, Zhang X. Somatosensory neuron types identified by high-coverage single-cell RNA-sequencing and functional heterogeneity. Cell Res 2015; 26:83-102. [PMID: 26691752 DOI: 10.1038/cr.2015.149] [Citation(s) in RCA: 268] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 11/04/2015] [Accepted: 12/01/2015] [Indexed: 11/09/2022] Open
Abstract
Sensory neurons are distinguished by distinct signaling networks and receptive characteristics. Thus, sensory neuron types can be defined by linking transcriptome-based neuron typing with the sensory phenotypes. Here we classify somatosensory neurons of the mouse dorsal root ganglion (DRG) by high-coverage single-cell RNA-sequencing (10 950 ± 1 218 genes per neuron) and neuron size-based hierarchical clustering. Moreover, single DRG neurons responding to cutaneous stimuli are recorded using an in vivo whole-cell patch clamp technique and classified by neuron-type genetic markers. Small diameter DRG neurons are classified into one type of low-threshold mechanoreceptor and five types of mechanoheat nociceptors (MHNs). Each of the MHN types is further categorized into two subtypes. Large DRG neurons are categorized into four types, including neurexophilin 1-expressing MHNs and mechanical nociceptors (MNs) expressing BAI1-associated protein 2-like 1 (Baiap2l1). Mechanoreceptors expressing trafficking protein particle complex 3-like and Baiap2l1-marked MNs are subdivided into two subtypes each. These results provide a new system for cataloging somatosensory neurons and their transcriptome databases.
Collapse
Affiliation(s)
- Chang-Lin Li
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 20031, China
| | - Kai-Cheng Li
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 20031, China
| | - Dan Wu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 20031, China
| | - Yan Chen
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 20031, China
| | - Hao Luo
- School of Life Science and Technology, ShanghaiTec University, Shanghai 200031, China
| | - Jing-Rong Zhao
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 20031, China
| | - Sa-Shuang Wang
- School of Life Science and Technology, ShanghaiTec University, Shanghai 200031, China
| | - Ming-Ming Sun
- National Engineering Center for Biochip at Shanghai, Shanghai, China
| | - Ying-Jin Lu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 20031, China
| | - Yan-Qing Zhong
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 20031, China
| | - Xu-Ye Hu
- Shanghai Clinical Center, Chinese Academy of Sciences/XuHui Central Hospital, Shanghai, China
| | - Rui Hou
- National Engineering Center for Biochip at Shanghai, Shanghai, China
| | - Bei-Bei Zhou
- National Engineering Center for Biochip at Shanghai, Shanghai, China
| | - Lan Bao
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences.,School of Life Science and Technology, ShanghaiTec University, Shanghai 200031, China
| | - Hua-Sheng Xiao
- National Engineering Center for Biochip at Shanghai, Shanghai, China
| | - Xu Zhang
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 20031, China.,School of Life Science and Technology, ShanghaiTec University, Shanghai 200031, China
| |
Collapse
|
20
|
Fukuoka T, Noguchi K. A potential anti-allodynic mechanism of GDNF following L5 spinal nerve ligation; Mitigation of NPY up-regulation in the touch sense pathway. Neuroscience 2015. [PMID: 26215916 DOI: 10.1016/j.neuroscience.2015.07.059] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Intrathecal delivery of glial cell line-derived neurotrophic factor (GDNF) reverses mechanical allodynia after 5th lumbar (L5) spinal nerve ligation (SNL). However, the molecular mechanism behind this process is not fully understood. Following sciatic nerve injury, primary afferent neurons in the injured dorsal root ganglion (DRG) begin to express neuropeptide Y (NPY) that is absent in normal DRG. The aim of the current study was to determine the relationship of this de novo expression of NPY and the anti-allodynic effect of GDNF. Following L5 SNL, 73% of neurons began to express NPY mRNA in the ipsilateral L5 DRG and robust NPY-immunoreactive fibers appeared in the ipsilateral GN where the touch-sense mediating A-fiber primary afferents from the hindpaw terminate. Seven-daylong intrathecal infusion of GDNF at the L5 DRG level, starting on day three when mechanical allodynia had fully developed, reversed once-established these changes. The GN neurons normally expressed NPY Y1 receptor, but not Y2, Y4, or Y5 receptors, and L5 SNL did not change the expression pattern. Bolus intracisternal injection of BIBP3226, a Y1 receptor antagonist, dose-dependently reversed mechanical allodynia. We demonstrated that GDNF reversed once-established mechanical allodynia as well as NPY induction in the touch-sense processing pathway. NPY could facilitate touch-sense processing by Y1 receptor in the gracile nucleus after peripheral nerve injury. GDNF may exert anti-allodynic effects through mitigation of this NPY up-regulation. The effectiveness of delayed treatment further indicates the therapeutic potential of GDNF on neuropathic pain.
Collapse
Affiliation(s)
- T Fukuoka
- Department of Anatomy & Neuroscience, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan; Pain Mechanism Research Group, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan.
| | - K Noguchi
- Department of Anatomy & Neuroscience, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| |
Collapse
|
21
|
Magnussen C, Hung SP, Ribeiro-da-Silva A. Novel expression pattern of neuropeptide Y immunoreactivity in the peripheral nervous system in a rat model of neuropathic pain. Mol Pain 2015; 11:31. [PMID: 26012590 PMCID: PMC4449610 DOI: 10.1186/s12990-015-0029-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 05/20/2015] [Indexed: 12/26/2022] Open
Abstract
Background Neuropeptide Y (NPY) has been implicated in the modulation of pain. Under normal conditions, NPY is found in interneurons in the dorsal horn of the spinal cord and in sympathetic postganglionic neurons but is absent from the cell bodies of sensory neurons. Following peripheral nerve injury NPY is dramatically upregulated in the sensory ganglia. How NPY expression is altered in the peripheral nervous system, distal to a site of nerve lesion, remains unknown. To address this question, NPY expression was investigated using immunohistochemistry at the level of the trigeminal ganglion, the mental nerve and in the skin of the lower lip in relation to markers of sensory and sympathetic fibers in a rat model of trigeminal neuropathic pain. Results At 2 and 6 weeks after chronic constriction injury (CCI) of the mental nerve, de novo expression of NPY was seen in the trigeminal ganglia, in axons in the mental nerve, and in fibers in the upper dermis of the skin. In lesioned animals, NPY immunoreactivity was expressed primarily by large diameter mental nerve sensory neurons retrogradely labelled with Fluorogold. Many axons transported this de novo NPY to the periphery as NPY-immunoreactive (IR) fibers were seen in the mental nerve both proximal and distal to the CCI. Some of these NPY-IR axons co-expressed Neurofilament 200 (NF200), a marker for myelinated sensory fibers, and occasionally colocalization was seen in their terminals in the skin. Peptidergic and non-peptidergic C fibers expressing calcitonin gene-related peptide (CGRP) or binding isolectin B4 (IB4), respectively, never expressed NPY. CCI caused a significant de novo sprouting of sympathetic fibers into the upper dermis of the skin, and most, but not all of these fibers, expressed NPY. Conclusions This is the first study to provide a comprehensive description of changes in NPY expression in the periphery after nerve injury. Novel expression of NPY in the skin comes mostly from sprouted sympathetic fibers. This information is fundamental in order to understand where endogenous NPY is expressed, and how it might be acting to modulate pain in the periphery.
Collapse
Affiliation(s)
- Claire Magnussen
- Department of Pharmacology and Therapeutics, McGill University, McIntyre Medical Building, 3655 Promenade Sir William Osler, Room 1215, Montreal, Quebec, H3G 1Y6, Canada. .,Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, H3A 0G1, Canada.
| | - Shih-Ping Hung
- Department of Pharmacology and Therapeutics, McGill University, McIntyre Medical Building, 3655 Promenade Sir William Osler, Room 1215, Montreal, Quebec, H3G 1Y6, Canada.
| | - Alfredo Ribeiro-da-Silva
- Department of Pharmacology and Therapeutics, McGill University, McIntyre Medical Building, 3655 Promenade Sir William Osler, Room 1215, Montreal, Quebec, H3G 1Y6, Canada. .,Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, H3A 0G1, Canada. .,Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, H3A 0C7, Canada.
| |
Collapse
|
22
|
de Lartigue G, Ronveaux CC, Raybould HE. Deletion of leptin signaling in vagal afferent neurons results in hyperphagia and obesity. Mol Metab 2014; 3:595-607. [PMID: 25161883 PMCID: PMC4142400 DOI: 10.1016/j.molmet.2014.06.003] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 06/18/2014] [Accepted: 06/21/2014] [Indexed: 12/15/2022] Open
Abstract
The vagal afferent pathway senses hormones released from the gut in response to nutritional cues and relays these signals to the brain. We tested the hypothesis that leptin resistance in vagal afferent neurons (VAN) is responsible for the onset of hyperphagia by developing a novel conditional knockout mouse to delete leptin receptor selectively in sensory neurons (Nav1.8/LepR (fl/fl) mice). Chow fed Nav1.8/LepR (fl/fl) mice weighed significantly more and had increased adiposity compared with wildtype mice. Cumulative food intake, meal size, and meal duration in the dark phase were increased in Nav1.8/LepR (fl/fl) mice; energy expenditure was unaltered. Reduced satiation in Nav1.8/LepR (fl/fl) mice is in part due to reduced sensitivity of VAN to CCK and the subsequent loss of VAN plasticity. Crucially Nav1.8/LepR (l/fl) mice did not gain further weight in response to a high fat diet. We conclude that disruption of leptin signaling in VAN is sufficient and necessary to promote hyperphagia and obesity.
Collapse
Affiliation(s)
- Guillaume de Lartigue
- Department of Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, 1 Shields Ave, Davis, CA 95616, USA
| | - Charlotte C Ronveaux
- Department of Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, 1 Shields Ave, Davis, CA 95616, USA
| | - Helen E Raybould
- Department of Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, 1 Shields Ave, Davis, CA 95616, USA
| |
Collapse
|
23
|
Sensory innervation of the dorsal longitudinal ligament and the meninges in the lumbar spine of the dog. Histochem Cell Biol 2014; 142:433-47. [PMID: 24748503 DOI: 10.1007/s00418-014-1218-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2014] [Indexed: 10/25/2022]
Abstract
Although intervertebral disc herniation is a well-known disease in dogs, pain management for this condition has remained a challenge. The goal of the present study is to address the lack of information regarding the innervation of anatomical structures within the canine vertebral canal. Immunolabeling was performed with antibodies against protein gene product 9.5, Tuj-1 (neuron-specific class III β-tubulin), calcitonin gene-related peptide, and neuropeptide Y in combination with the lectin from Lycopersicon esculentum as a marker for blood vessels. Staining was indicative of both sensory and sympathetic fibers. Innervation density was the highest in lateral areas, intermediate in dorsal areas, and the lowest in ventral areas. In the dorsal longitudinal ligament (DLL), the highest innervation density was observed in the lateral regions. Innervation was lower at mid-vertebral levels than at intervertebral levels. The presence of sensory and sympathetic fibers in the canine dura and DLL suggests that pain may originate from both these structures. Due to these regional differences in sensory innervation patterns, trauma to intervertebral DLL and lateral dura is expected to be particularly painful. The results ought to provide a better basis for the assessment of medicinal and surgical procedures.
Collapse
|
24
|
Holzer P, Reichmann F, Farzi A. Neuropeptide Y, peptide YY and pancreatic polypeptide in the gut-brain axis. Neuropeptides 2012; 46:261-74. [PMID: 22979996 PMCID: PMC3516703 DOI: 10.1016/j.npep.2012.08.005] [Citation(s) in RCA: 342] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 08/04/2012] [Accepted: 08/09/2012] [Indexed: 02/06/2023]
Abstract
The gut-brain axis refers to the bidirectional communication between the gut and the brain. Four information carriers (vagal and spinal afferent neurons, immune mediators such as cytokines, gut hormones and gut microbiota-derived signalling molecules) transmit information from the gut to the brain, while autonomic neurons and neuroendocrine factors carry outputs from the brain to the gut. The members of the neuropeptide Y (NPY) family of biologically active peptides, NPY, peptide YY (PYY) and pancreatic polypeptide (PP), are expressed by cell systems at distinct levels of the gut-brain axis. PYY and PP are exclusively expressed by endocrine cells of the digestive system, whereas NPY is found at all levels of the gut-brain and brain-gut axis. The major systems expressing NPY comprise enteric neurons, primary afferent neurons, several neuronal pathways throughout the brain and sympathetic neurons. In the digestive tract, NPY and PYY inhibit gastrointestinal motility and electrolyte secretion and in this way modify the input to the brain. PYY is also influenced by the intestinal microbiota, and NPY exerts, via stimulation of Y1 receptors, a proinflammatory action. Furthermore, the NPY system protects against distinct behavioural disturbances caused by peripheral immune challenge, ameliorating the acute sickness response and preventing long-term depression. At the level of the afferent system, NPY inhibits nociceptive input from the periphery to the spinal cord and brainstem. In the brain, NPY and its receptors (Y1, Y2, Y4, Y5) play important roles in regulating food intake, energy homeostasis, anxiety, mood and stress resilience. In addition, PP and PYY signal to the brain to attenuate food intake, anxiety and depression-related behaviour. These findings underscore the important role of the NPY-Y receptor system at several levels of the gut-brain axis in which NPY, PYY and PP operate both as neural and endocrine messengers.
Collapse
Affiliation(s)
- Peter Holzer
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, A-8010 Graz, Austria.
| | | | | |
Collapse
|
25
|
Kostić S, Puljak L, Sapunar D. Attenuation of pain-related behaviour evoked by carrageenan injection through blockade of neuropeptide Y Y1 and Y2 receptors. Eur J Pain 2012; 17:493-504. [DOI: 10.1002/j.1532-2149.2012.00218.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2012] [Indexed: 01/01/2023]
Affiliation(s)
- S. Kostić
- Laboratory for Pain Research; Department of Anatomy, Histology and Embryology; University of Split School of Medicine; Croatia
| | - L. Puljak
- Laboratory for Pain Research; Department of Anatomy, Histology and Embryology; University of Split School of Medicine; Croatia
| | - D. Sapunar
- Laboratory for Pain Research; Department of Anatomy, Histology and Embryology; University of Split School of Medicine; Croatia
| |
Collapse
|
26
|
Singla S, Ding Z, Mehta JL. Arterial wall-axon reflex: a potential player in atherosclerosis. Ther Adv Cardiovasc Dis 2012; 6:181-4. [PMID: 23045192 DOI: 10.1177/1753944712460421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
27
|
Abstract
Here we used an array-based differential screen to uncover the expression of the neuropeptide neuromedin B (NMB) in the trigeminal ganglia of mice. Double-labeling experiments reveal NMB is expressed in a subset of sensory neurons that colabel with calcitonin gene-related peptide and TRPV1 suggestive of a role for NMB in nociception. Indeed, administration of NMB antagonist greatly attenuates edema and nerve sensitization following stimulation of peripheral nerves with mustard oil, demonstrating that NMB contributes to neurogenic inflammation. Moreover, direct injection of NMB causes local swelling and nociceptive sensitization. Interestingly, we also find that the receptor for NMB is expressed in interneurons in the superficial layers of the dorsal horn. We used NMB-saporin to specifically eliminate NMBR-expressing neurons and determined they are required in responses to noxious heat, but not for reaction to mechanical and pruritic stimuli. Thus, NMB may be a neurotransmitter that is selectively involved in the perception of thermal stimuli.
Collapse
|
28
|
Page AJ, Symonds E, Peiris M, Blackshaw LA, Young RL. Peripheral neural targets in obesity. Br J Pharmacol 2012; 166:1537-58. [PMID: 22432806 PMCID: PMC3419899 DOI: 10.1111/j.1476-5381.2012.01951.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Revised: 02/20/2012] [Accepted: 02/22/2012] [Indexed: 12/15/2022] Open
Abstract
Interest in pharmacological treatments for obesity that act in the brain to reduce appetite has increased exponentially over recent years, but failures of clinical trials and withdrawals due to adverse effects have so far precluded any success. Treatments that do not act within the brain are, in contrast, a neglected area of research and development. This is despite the fact that a vast wealth of molecular mechanisms exists within the gut epithelium and vagal afferent system that could be manipulated to increase satiety. Here we discuss mechano- and chemosensory pathways from the gut involved in appetite suppression, and distinguish between gastric and intestinal vagal afferent pathways in terms of their basic physiology and activation by enteroendocrine factors. Gastric bypass surgery makes use of this system by exposing areas of the intestine to greater nutrient loads resulting in greater satiety hormone release and reduced food intake. A non-surgical approach to this system is preferable for many reasons. This review details where the opportunities may lie for such approaches by describing nutrient-sensing mechanisms throughout the gastrointestinal tract.
Collapse
Affiliation(s)
- Amanda J Page
- Nerve-Gut Research Laboratory, Discipline of Medicine, South Australia, Australia
| | | | | | | | | |
Collapse
|
29
|
Wang D, Chen T, Gao Y, Quirion R, Hong Y. Inhibition of SNL-induced upregulation of CGRP and NPY in the spinal cord and dorsal root ganglia by the 5-HT(2A) receptor antagonist ketanserin in rats. Pharmacol Biochem Behav 2012; 101:379-86. [PMID: 22342663 DOI: 10.1016/j.pbb.2012.02.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Revised: 02/01/2012] [Accepted: 02/05/2012] [Indexed: 12/11/2022]
Abstract
Our previous study has demonstrated that topical and systemic administration of the 5-HT(2A) receptor antagonist ketanserin attenuates neuropathic pain. To explore the mechanisms involved, we examined whether ketanserin reversed the plasticity changes associated with calcitonin gene-related peptides (CGRP) and neuropeptide Y (NPY) which may reflect distinct mechanisms: involvement and compensatory protection. Behavioral responses to thermal and tactile stimuli after spinal nerve ligation (SNL) at L5 demonstrated neuropathic pain and its attenuation in the vehicle- and ketanserin-treated groups, respectively. SNL surgery induced an increase in CGRP and NPY immunoreactivity (IR) in laminae I-II of the spinal cord. L5 SNL produced an expression of NPY-IR in large, medium and small diameter neurons in dorsal root ganglion (DRG) only at L5, but not adjacent L4 and L6. Daily injection of ketanserin (0.3 mg/kg, s.c.) for two weeks suppressed the increase in CGRP-IR and NPY-IR in the spinal cord or DRG. The present study demonstrated that: (1) the expression of CGRP was enhanced in the spinal dorsal horn and NPY was expressed in the DRG containing injured neurons, but not in the adjacent DRG containing intact neurons, following L5 SNL; (2) the maladaptive changes in CGRP and NPY expression in the spinal cord and DRG mediated the bioactivity of 5-HT/5-HT(2A) receptors in neuropathic pain and (3) the blockade of 5-HT(2A) receptors by ketanserin reversed the evoked upregulation of both CGRP and NPY in the spinal cord and DRG contributing to the inhibition of neuropathic pain.
Collapse
Affiliation(s)
- Dongmei Wang
- Provincial Key Laboratory of Developmental Biology and Neuroscience, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian, 350108, People's Republic of China
| | | | | | | | | |
Collapse
|
30
|
Steinert RE, Beglinger C. Nutrient sensing in the gut: interactions between chemosensory cells, visceral afferents and the secretion of satiation peptides. Physiol Behav 2011; 105:62-70. [DOI: 10.1016/j.physbeh.2011.02.039] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 02/24/2011] [Accepted: 02/25/2011] [Indexed: 01/01/2023]
|
31
|
Kaczyńska K, Szereda-Przestaszewska M. Activation of neuropeptide Y(2) receptors exerts an excitatory action on cardio-respiratory variables in anaesthetized rats. Neuropeptides 2011; 45:281-6. [PMID: 21658765 DOI: 10.1016/j.npep.2011.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Revised: 05/17/2011] [Accepted: 05/18/2011] [Indexed: 12/26/2022]
Abstract
The respiratory effects of stimulation of NPYY(2) receptors were studied in spontaneously breathing rats that were either (i) neurally intact and subsequently bilaterally vagotomized in the neck, or (ii) neurally intact and subjected to supranodosal vagotomy or (iii) neurally intact treated with pharmacological blockade of NPY(1-2) receptors. Before neural interventions an intravenous (iv) bolus of the NPYY(2) receptor agonist NPY 13-36 (10 μg/kg) increased breathing rate, tidal volume and mean arterial blood pressure (MAP). Section of the midcervical vagi abrogated NPY 13-36-evoked increase in respiratory rate but had no effect on augmented tidal volume, minute ventilation and blood pressure. Supranodosal vagotomy prevented the increase in tidal volume and slightly reduced the pressor response. Blockade of NPYY(2) receptor with intravenous doses of BIIE 0246 eliminated cardio-respiratory effects of NPY 13-36 injection. BMS 193885 - an antagonist of NPYY(1) receptor-was not effective in abrogating cardio-respiratory response. The present study showed that (i) NPY 13-36 induced stimulation of breathing results from activation of NPYY(2) receptors associated with pulmonary vagal afferentation; (ii) the increase in the frequency of breathing is mediated by midcervical vagi and augmentation of tidal volume relies on the intact supranodosal trunks (iii) the pressor response results from the excitation of NPYY(2) receptors outside of the vagal pathway.
Collapse
Affiliation(s)
- Katarzyna Kaczyńska
- Laboratory of Respiratory Reflexes, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.
| | | |
Collapse
|
32
|
Sapunar D, Vukojević K, Kostić S, Puljak L. Attenuation of pain-related behavior evoked by injury through blockade of neuropeptide Y Y2 receptor. Pain 2011; 152:1173-1181. [PMID: 21376464 DOI: 10.1016/j.pain.2011.01.045] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Revised: 01/17/2011] [Accepted: 01/24/2011] [Indexed: 01/02/2023]
Abstract
Neuropeptide Y (NPY) has an important but still insufficiently defined role in pain modulation. We therefore examined the ability of NPY to modulate experimentally induced neuropathic pain by injecting it directly into dorsal root ganglion (DRG) immediately following spinal nerve ligation (SNL) injury. We have found that this application exacerbates pain-related behavior induced by SNL in a modality-specific fashion. When saline was injected after SNL, the expected increase in hyperalgesia responses to needle stimulation was present on the 8th postoperative day. When we injected NPY, hyperalgesic responses were increased in a manner similar to the SNL/saline group. To characterize NPY action, specific Y1 and Y2 antagonists were also delivered directly to DRG, which revealed that behavioral actions of NPY were abolished by Y2 receptor antagonist. We tested whether NPY effects were the result of its role in immunity by immunohistochemical staining for glial fibrillary acidic protein, in order to identify activation of DRG satellite cells and dorsal horn astrocytes. Exacerbation of pain-related behavior following NPY injection was accompanied by astrocyte activation in ipsilateral dorsal horn and with satellite cells activation in the DRG proximal to injury. This activation was reduced following Y2 receptor antagonist application. These findings indicate an important link between pain-related behavior and neuroimmune activation by NPY through its Y2 receptor.
Collapse
Affiliation(s)
- Damir Sapunar
- Laboratory for Pain Research, Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2, Split 21000, Croatia
| | | | | | | |
Collapse
|
33
|
Dockray GJ, Burdyga G. Plasticity in vagal afferent neurones during feeding and fasting: mechanisms and significance. Acta Physiol (Oxf) 2011; 201:313-21. [PMID: 21062423 DOI: 10.1111/j.1748-1716.2010.02219.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The ingestion of food activates mechanisms leading to inhibition of food intake and gastric emptying mediated by the release of regulatory peptides, for example cholecystokinin (CCK), and lipid amides, e.g. oleylethanolamide from the gut. In addition, there are both peptides (e.g. ghrelin) and lipid amides (e.g. anandamide) that appear to signal the absence of food in the gut and that are associated with the stimulation of food intake. Vagal afferent neurones are a common target for both types of signal. Remarkably, the neurochemical phenotype of these neurones itself depends on nutritional status. CCK acting at CCK1 receptors on vagal afferent neurones stimulates expression in these neurones of Y2-receptors and the neuropeptide CART, both of which are associated with the inhibition of food intake. Conversely, in fasted rats when plasma CCK is low, these neurones express cannabinoid (CB)-1 and melanin concentrating hormone (MCH)-1 receptors, and MCH, and this is inhibited by exogenous CCK or endogenous CCK released by refeeding. The stimulation of CART expression by CCK is mediated by the activation of CREB and EGR1; ghrelin inhibits the action of CCK by promoting nuclear exclusion of CREB and leptin potentiates the action of CCK by the stimulation of EGR1 expression. Vagal afferent neurones therefore constitute a level of integration outside the CNS for nutrient-derived signals that control energy intake and that are capable of encoding recent nutrient ingestion.
Collapse
Affiliation(s)
- G J Dockray
- Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, Crown Street, Liverpool, UK.
| | | |
Collapse
|
34
|
Zhang YP, Fu ES, Sagen J, Levitt RC, Candiotti KA, Bethea JR, Brambilla R. Glial NF-κB inhibition alters neuropeptide expression after sciatic nerve injury in mice. Brain Res 2011; 1385:38-46. [PMID: 21352816 DOI: 10.1016/j.brainres.2011.02.055] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Revised: 01/25/2011] [Accepted: 02/16/2011] [Indexed: 01/06/2023]
Abstract
We utilized a transgenic mouse model where nuclear factor kappa B (NF-κB) is selectively inhibited in glial fibrillary acidic protein (GFAP) expressing cells. The transgene, GFAP-IκBα-dn, overexpresses a dominant negative form of the inhibitor of NF-κB (IκBα) under the control of the GFAP promoter. In the present work, we sought to understand the impact of glial NF-κB inhibition on the expression of pain mediating sensory neuropeptides galanin and calcitonin gene related peptide (CGRP) in a model of neuropathic pain in mice. Chronic constriction injury (CCI) of the left sciatic nerve was performed on wild type (WT) and GFAP-IκBα-dn transgenic mice. RT-PCR and immunohistological staining were performed in sciatic nerve and/or L4-L5 DRG tissue for galanin, CGRP and macrophage marker CD11b. GFAP-IκBα-dn mice had less mechanical and thermal hyperalgesia compared to WT mice post-CCI. After CCI, we observed galanin upregulation in DRG and sciatic nerve, which was less in GFAP-IκBα-dn mice. CGRP gene expression in the DRG increased transiently on day 1 post-CCI in WT but not in GFAP-IκBα-dn mice, and no evidence of CGRP upregulation in sciatic nerve post-CCI was found. After CCI, upregulation of CD11b in sciatic nerve was less in GFAP-IκBα-dn mice compared to WT mice, indicative of less macrophage infiltration. Our results showed that glial NF-κB inhibition reduces galanin and CGRP expression, which are neuropeptides that correlate with pain behavior and inflammation after peripheral nerve injury.
Collapse
Affiliation(s)
- Yan Ping Zhang
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Steinert RE, Poller B, Castelli MC, Drewe J, Beglinger C. Oral administration of glucagon-like peptide 1 or peptide YY 3-36 affects food intake in healthy male subjects. Am J Clin Nutr 2010; 92:810-7. [PMID: 20720259 DOI: 10.3945/ajcn.2010.29663] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Peripheral infusion of glucagon-like peptide 1 (GLP-1) or peptide YY 3-36 (PYY3-36) reduces food intake in healthy, obese, and diabetic subjects. In vivo, both peptides are cosecreted from intestinal L cells; GLP-1 is subject to rapid breakdown by dipeptidyl peptidase IV, and together with PYY3-36 it is likely to be degraded in the liver before entering the systemic circulation. The largest concentrations are observed in the splanchnic blood rather than in the systemic circulation. OBJECTIVE In contrast with peripheral infusion, oral delivery of sodium N-[8-(2-hydroxybenzoyl) amino] caprylate (SNAC) mimics endogenous secretion. We aimed to investigate how this affects food intake. DESIGN Twelve healthy male subjects were studied in a randomized, double-blind, placebo-controlled, 4-way crossover trial. Each subject received in random order 2.0 mg GLP-1, 1.0 mg PYY3-36, or 2.0 mg GLP-1 plus 1.0 mg PYY3-36; the peptides were mixed with SNAC. The placebo treatment was the delivery agent alone. Food intake during an ad libitum test meal was measured. RESULTS Both peptides were rapidly absorbed from the gut, leading to plasma concentrations several times higher than those in response to a normal meal. GLP-1 alone, but not PYY3-36, reduced total energy intake significantly, with marked effects on glucose homeostasis. Coadministration of both peptides reduced total energy intake by 21.5% and fullness at meal onset (P < 0.05) but not total 24-h energy intake. CONCLUSION The results show a marked effect of orally administered GLP-1 and PYY3-36 on appetite by showing enhanced fullness at meal onset and reduced energy intake. This trial was registered at clinicaltrials.gov as NCT00822705.
Collapse
Affiliation(s)
- Robert E Steinert
- Clinical Research Center, Department of Biomedicine, University Hospital, Basel, Switzerland
| | | | | | | | | |
Collapse
|
36
|
Inhibition of inflammatory pain by activating B-type natriuretic peptide signal pathway in nociceptive sensory neurons. J Neurosci 2010; 30:10927-38. [PMID: 20702721 DOI: 10.1523/jneurosci.0657-10.2010] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
B-type natriuretic peptide (BNP) has been known to be secreted from cardiac myocytes and activate its receptor, natriuretic peptide receptor-A (NPR-A), to reduce ventricular fibrosis. However, the function of BNP/NPR-A pathway in the somatic sensory system has been unknown. In the present study, we report a novel function of BNP in pain modulation. Using microarray and immunoblot analyses, we found that BNP and NPR-A were expressed in the dorsal root ganglion (DRG) of rats and upregulated after intraplantar injection of complete Freund's adjuvant (CFA). Immunohistochemistry showed that BNP was expressed in calcitonin gene-related peptide (CGRP)-containing small neurons and IB4 (isolectin B4)-positive neurons, whereas NPR-A was present in CGRP-containing neurons. Application of BNP reduced the firing frequency of small DRG neurons in the presence of glutamate through opening large-conductance Ca2+-activated K+ channels (BKCa channels). Furthermore, intrathecal injection of BNP yielded inhibitory effects on formalin-induced flinching behavior and CFA-induced thermal hyperalgesia in rats. Blockade of BNP signaling by BNP antibodies or cGMP-dependent protein kinase (PKG) inhibitor KT5823 [(9S,10R,12R)-2,3,9,10,11,12-hexahydro-10-methoxy-2,9-dimethyl-1-oxo-9,12-epoxy-1H-diindolo[1,2,3-fg:3',2',1'-kl]pyrrolo[3,4-i][1,6]benzodiazocine-10-carboxylic acid methyl ester] impaired the recovery from CFA-induced thermal hyperalgesia. Thus, BNP negatively regulates nociceptive transmission through presynaptic receptor NPR-A, and activation of the BNP/NPR-A/PKG/BKCa channel pathway in nociceptive afferent neurons could be a potential strategy for inflammatory pain therapy.
Collapse
|
37
|
Lee NJ, Allison S, Enriquez RF, Sainsbury A, Herzog H, Baldock PA. Y2 and Y4 Receptor Signalling Attenuates the Skeletal Response of Central NPY. J Mol Neurosci 2010; 43:123-31. [DOI: 10.1007/s12031-010-9423-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Accepted: 07/01/2010] [Indexed: 12/30/2022]
|
38
|
Baraboi ED, Michel C, Smith P, Thibaudeau K, Ferguson AV, Richard D. Effects of albumin-conjugated PYY on food intake: the respective roles of the circumventricular organs and vagus nerve. Eur J Neurosci 2010; 32:826-39. [DOI: 10.1111/j.1460-9568.2010.07318.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
39
|
Dockray GJ. Cholecystokinin and gut-brain signalling. ACTA ACUST UNITED AC 2009; 155:6-10. [PMID: 19345244 DOI: 10.1016/j.regpep.2009.03.015] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2009] [Accepted: 03/25/2009] [Indexed: 01/04/2023]
Abstract
Enteroendocrine cells of the gastrointestinal tract act as a luminal surveillance system responding to either the presence or absence of food in the gut lumen. Collectively, their secretory products regulate the course of digestion and determine the delivery of nutrient to the gut by controlling food intake. Afferent neurons of the vagus nerve are an important target of gut hormones, particularly for control of food intake. The intestinal hormone cholecystokinin (CCK) stimulates vagal afferent neuron discharge and also controls the expression of both G-protein coupled receptors and peptide neurotransmitters in these neurons. When plasma CCK concentrations are low, for example in fasting, vagal afferent neurons express cannabinoid CB1 and melanin concentrating hormone (MCH)-1 receptors, both of which are associated with stimulation of food intake. Post-prandial release of CCK rapidly down-regulates the expression of both receptors but stimulates the expression of Y2 receptors in neurons projecting to the stomach. In fasting, there is also increased expression in these neurons of the appetite-stimulating neuropeptide transmitter MCH, and depressed expression of the satiety-peptide cocaine and amphetamine regulated transcript (CART). Secretion of CCK decreases expression of MCH and increases expression of CART. The neurochemical phenotype of vagal afferent neurons therefore encodes whether or not there has been nutrient ingestion over the previous period. At low plasma concentrations of CCK vagal afferent neurons exhibit increased capacity for appetite-stimulation, while post-prandial concentrations of CCK lead to enhanced capacity for satiety signalling. A gatekeeper function can therefore be attributed to CCK in that its presence or absence influences the capacity of vagal afferent neurons to respond to other neurohormonal signals.
Collapse
Affiliation(s)
- Graham J Dockray
- Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, Crown St, Liverpool L69 3BX, UK.
| |
Collapse
|
40
|
Coronel MF, Musolino PL, Brumovsky PR, Hökfelt T, Villar MJ. Bone marrow stromal cells attenuate injury-induced changes in galanin, NPY and NPY Y1-receptor expression after a sciatic nerve constriction. Neuropeptides 2009; 43:125-32. [PMID: 19168218 DOI: 10.1016/j.npep.2008.12.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2008] [Revised: 12/11/2008] [Accepted: 12/12/2008] [Indexed: 12/21/2022]
Abstract
Single ligature nerve constriction (SLNC) of the rat sciatic nerve triggers neuropathic pain-related behaviors and induces changes in neuropeptide expression in primary afferent neurons. Bone marrow stromal cells (MSCs) injected into the lumbar 4 (L4) dorsal root ganglia (DRGs) of animals subjected to a sciatic nerve SLNC selectively migrate to the other ipsilateral lumbar DRGs (L3, L5 and L6) and prevent mechanical and thermal allodynia. In this study, we have evaluated the effect of MSC administration on the expression of the neuropeptides galanin and NPY, as well as the NPY Y(1)-receptor (Y(1)R) in DRG neurons. Animals were subjected to a sciatic nerve SLNC either alone or followed by the administration of MSCs, phosphate-buffered saline (PBS) or bone marrow non-adherent mononuclear cells (BNMCs), directly into the ipsilateral L4 DRG. Seven days after injury, the ipsilateral and contralateral L4-5 DRGs were dissected out and processed for standard immunohistochemistry, using specific antibodies. As previously reported, SLNC induced an ipsilateral increase in the number of galanin and NPY immunoreactive neurons and a decrease in Y(1)R-positive DRG neurons. The intraganglionic injection of PBS or BNMCs did not modify this pattern of expression. In contrast, MSC administration partially prevented the injury-induced changes in galanin, NPY and Y(1)R expression. The large number of Y(1)R-immunoreactive neurons together with high levels of NPY expression in animals injected with MSCs could explain, at least in part, the analgesic effects exerted by these cells. Our results support MSC participation in the modulation of neuropathic pain and give insight into one of the possible mechanisms involved.
Collapse
Affiliation(s)
- M F Coronel
- School of Biomedical Sciences, Austral University, Buenos Aires, Argentina.
| | | | | | | | | |
Collapse
|
41
|
Dockray GJ. The versatility of the vagus. Physiol Behav 2009; 97:531-6. [PMID: 19419683 DOI: 10.1016/j.physbeh.2009.01.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Revised: 01/13/2009] [Accepted: 01/14/2009] [Indexed: 12/24/2022]
Abstract
The gut is one of several organs contributing to the peripheral signalling network that controls food intake. Afferent neurons of the vagus nerve provide an important pathway for gut signals that act by triggering ascending pathways from the brain stem to hypothalamus. Recent work indicates the existence of mechanisms operating at the level of vagal afferent neurons to modulate the effect of gastrointestinal satiety signals. Thus, the well known satiety hormone cholecystokinin (CCK) not only stimulates the discharge of these neurons but also controls their expression of both G-protein coupled receptors and peptide neurotransmitters known to influence food intake. When plasma CCK concentrations are low e.g. in fasting, the expression by vagal afferent neurons of cannabinoid (CB)-1 and melanin concentrating hormone (MCH)-1 receptors is increased. Release of CCK by feeding leads to a rapid down-regulation of expression of both receptors and to increased expression of Y2 receptors. In fasting, there is also increased expression in these neurons of the appetite-stimulating neuropeptide transmitter MCH, and depressed expression of the satiety-peptide cocaine and amphetamine regulated transcript (CARTp); endogenous CCK decreases MCH expression and stimulates CART expression. The gastric orexigenic hormone ghrelin blocks these actions of CCK at least in part by excluding phosphoCREB from the nucleus. The data suggest that CCK acts as a gatekeeper to determine the capacity of other neuroendocrine signals to act via vagal afferent neurons to influence food intake.
Collapse
Affiliation(s)
- Graham J Dockray
- Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, Liverpool, UK.
| |
Collapse
|
42
|
Cholecystokinin regulates expression of Y2 receptors in vagal afferent neurons serving the stomach. J Neurosci 2008; 28:11583-92. [PMID: 18987194 DOI: 10.1523/jneurosci.2493-08.2008] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The intestinal hormones CCK and PYY3-36 inhibit gastric emptying and food intake via vagal afferent neurons. Here we report that CCK regulates the expression of Y2R, at which PYY3-36 acts. In nodose ganglia from rats fasted up to 48 h, there was a fivefold decrease of Y2R mRNA compared with rats fed ad libitum; Y2R mRNA in fasted rats was increased by administration of CCK, and by refeeding through a mechanism sensitive to the CCK1R antagonist lorglumide. Antibodies to Y2R revealed expression in both neurons and satellite cells; most of the former (89 +/- 4%) also expressed CCK1R. With fasting there was loss of Y2R immunoreactivity in CCK1R-expressing neurons many of which projected to the stomach, but not in satellite cells or neurons projecting to the ileum or proximal colon. Expression of a Y2R promoter-luciferase reporter (Y2R-luc) in cultured vagal afferent neurons was increased in response to CCK by 12.3 +/- 0.1-fold and by phorbol ester (16.2 +/- 0.4-fold); the response to both was abolished by the protein kinase C inhibitor Ro-32,0432. PYY3-36 stimulated CREB phosphorylation in rat nodose neurons after priming with CCK; in wild-type mice PYY3-36 increased Fos labeling in brainstem neurons but in mice null for CCK1R this response was abolished. Thus Y2R is expressed by functionally distinct subsets of nodose ganglion neurons projecting to the stomach and ileum/colon; in the former expression is dependent on stimulation by CCK, and there is evidence that PYY3-36 effects on vagal afferent neurons are CCK dependent.
Collapse
|
43
|
Abstract
Neuropeptide Y (NPY) is an important modulatory neuropeptide that regulates several physiological systems, including the activity of sensory neurons. We evaluated whether activation of the NPY Y1 receptor could modulate the activity of capsaicin-sensitive nociceptors in trigeminal ganglia and dental pulp. We tested this hypothesis by measuring capsaicin-stimulated calcitonin gene-related peptide release (CGRP) as a measure of nociceptor activity. Capsaicin-evoked CGRP release was inhibited by 50% (p < 0.05) in trigeminal ganglia and by 26% (p < 0.05) in dental pulp when tissues were pre-treated with [Leu(31),Pro(34)]NPY. The Y1 receptor was found to co-localize with the capsaicin receptor TRPV1 in trigeminal ganglia. These results demonstrate that activation of the Y1 receptor results in the inhibition of the activity of capsaicin-sensitive nociceptors in the trigeminal ganglia and dental pulp. These findings are relevant to the physiological modulation of dental nociceptors by endogenous NPY and demonstrate an important novel analgesic target for the treatment of dental pain.
Collapse
Affiliation(s)
- J L Gibbs
- Department of Endodontics, University of Texas Health Science Center in San Antonio, TX, USA.
| | | |
Collapse
|
44
|
Lenard NR, Berthoud HR. Central and peripheral regulation of food intake and physical activity: pathways and genes. Obesity (Silver Spring) 2008; 16 Suppl 3:S11-22. [PMID: 19190620 PMCID: PMC2687326 DOI: 10.1038/oby.2008.511] [Citation(s) in RCA: 222] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A changing environment and lifestyle on the background of evolutionary engraved and perinatally imprinted physiological response patterns is the foremost explanation for the current obesity epidemic. However, it is not clear what the mechanisms are by which the modern environment overrides the physiological controls of appetite and homeostatic body-weight regulation. Food intake and energy expenditure are controlled by complex, redundant, and distributed neural systems involving thousands of genes and reflecting the fundamental biological importance of adequate nutrient supply and energy balance. There has been much progress in identifying the important role of hypothalamus and caudal brainstem in the various hormonal and neural mechanisms by which the brain informs itself about availability of ingested and stored nutrients and, in turn, generates behavioral, autonomic, and endocrine output. Some of the genes involved in this "homeostatic" regulator are crucial for energy balance as manifested in the well-known monogenic obesity models. However, it can be clearly demonstrated that much larger portions of the nervous system of animals and humans, including the cortex, basal ganglia, and the limbic system, are concerned with the procurement of food as a basic and evolutionarily conserved survival mechanism to defend the lower limits of adiposity. By forming representations and reward expectancies through processes of learning and memory, these systems evolved to engage powerful emotions for guaranteed supply with, and ingestion of, beneficial foods from a sparse and often hostile environment. They are now simply overwhelmed with an abundance of food and food cues no longer contested by predators and interrupted by famines. The anatomy, chemistry, and functions of these elaborate neural systems and their interactions with the "homeostatic" regulator in the hypothalamus are poorly understood, and many of the genes involved are either unknown or not well characterized. This is regrettable because these systems are directly and primarily involved in the interactions of the modern environment and lifestyle with the human body. They are no less "physiological" than metabolic-regulatory mechanisms that have attracted most of the research during the past 15 years.
Collapse
Affiliation(s)
- Natalie R. Lenard
- Neurobiology of Nutrition Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | - Hans-Rudolf Berthoud
- Neurobiology of Nutrition Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| |
Collapse
|
45
|
Abstract
Food intake and energy expenditure are controlled by complex, redundant, and distributed neural systems that reflect the fundamental biological importance of adequate nutrient supply and energy balance. Much progress has been made in identifying the various hormonal and neural mechanisms by which the brain informs itself about availability of ingested and stored nutrients and, in turn, generates behavioral, autonomic, and endocrine output. While hypothalamus and caudal brainstem play crucial roles in this homeostatic function, areas in the cortex and limbic system are important for processing information regarding prior experience with food, reward, and emotion, as well as social and environmental context. Most vertebrates can store a considerable amount of energy as fat for later use, and this ability has now become one of the major health risks for many human populations. The predisposition to develop obesity can theoretically result from any pathological malfunction or lack of adaptation to changing environments of this highly complex system.
Collapse
Affiliation(s)
- Hans-Rudolf Berthoud
- Neurobiology of Nutrition Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
| | | |
Collapse
|
46
|
Parkinson JRC, Dhillo WS, Small CJ, Chaudhri OB, Bewick GA, Pritchard I, Moore S, Ghatei MA, Bloom SR. PYY3-36 injection in mice produces an acute anorexigenic effect followed by a delayed orexigenic effect not observed with other anorexigenic gut hormones. Am J Physiol Endocrinol Metab 2008; 294:E698-708. [PMID: 18285527 DOI: 10.1152/ajpendo.00405.2007] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Peptide YY (PYY) is secreted postprandially from the endocrine L cells of the gastrointestinal tract. PYY(3-36), the major circulating form of the peptide, is thought to reduce food intake in humans and rodents via high-affinity binding to the autoinhibitory neuropeptide Y (NPY) receptor within the arcuate nucleus. We studied the effect of early light-phase injection of PYY(3-36) on food intake in mice fasted for 0, 6, 12, 18, 24, and 30 h and show that PYY(3-36) produces an acute anorexigenic effect regardless of the duration of fasting. We also show evidence of a delayed orexigenic effect in ad libitum-fed mice injected with PYY(3-36) in the early light phase. This delayed orexigenic effect also occurs in mice administered a potent analog of PYY(3-36), d-Allo Ile(3) PYY(3-36), but not following injection of other anorectic agents (glucagon-like-peptide 1, oxyntomodulin, and lithium chloride). Early light-phase injection of PYY(3-36) to ad libitum-fed mice resulted in a trend toward increased levels of hypothalamic NPY and agouti-related peptide mRNA and a decrease in proopiomelanocortin mRNA at the beginning of the dark phase. Furthermore, plasma levels of ghrelin were increased significantly, and there was a trend toward decreased plasma PYY(3-36) levels at the beginning of the dark phase. These data indicate that PYY(3-36) injection results in an acute anorexigenic effect followed by a delayed orexigenic effect.
Collapse
Affiliation(s)
- James R C Parkinson
- Department of Metabolic Medicine, Hammersmith Hospital, Imperial College London, Du Cane Road, London, W12 ONN UK
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
The vagus nerve, food intake and obesity. ACTA ACUST UNITED AC 2008; 149:15-25. [PMID: 18482776 DOI: 10.1016/j.regpep.2007.08.024] [Citation(s) in RCA: 211] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2006] [Accepted: 08/08/2007] [Indexed: 01/01/2023]
Abstract
Food interacts with sensors all along the alimentary canal to provide the brain with information regarding its composition, energy content, and beneficial effect. Vagal afferents innervating the gastrointestinal tract, pancreas, and liver provide a rapid and discrete account of digestible food in the alimentary canal, as well as circulating and stored fuels, while vagal efferents, together with the sympathetic nervous system and hormonal mechanisms, codetermine the rate of nutrient absorption, partitioning, storage, and mobilization. Although vagal sensory mechanisms play a crucial role in the neural mechanism of satiation, there is little evidence suggesting a significant role in long-term energy homeostasis. However, increasing recognition of vagal involvement in the putative mechanisms making bariatric surgeries the most effective treatment for obesity should greatly stimulate future research to uncover the many details regarding the specific transduction mechanisms in the periphery and the inter- and intra-neuronal signaling cascades disseminating vagal information across the neuraxis.
Collapse
|
48
|
Chottová Dvoráková M, Wiegand S, Pesta M, Slavíková J, Grau V, Reischig J, Kuncová J, Kummer W. Expression of neuropeptide Y and its receptors Y1 and Y2 in the rat heart and its supplying autonomic and spinal sensory ganglia in experimentally induced diabetes. Neuroscience 2008; 151:1016-28. [DOI: 10.1016/j.neuroscience.2007.07.069] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2006] [Revised: 07/12/2007] [Accepted: 12/06/2007] [Indexed: 12/27/2022]
|
49
|
Hökfelt T, Brumovsky P, Shi T, Pedrazzini T, Villar M. NPY and pain as seen from the histochemical side. Peptides 2007; 28:365-72. [PMID: 17234301 DOI: 10.1016/j.peptides.2006.07.024] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2006] [Accepted: 07/20/2006] [Indexed: 01/10/2023]
Abstract
The expression of neuropeptide tyrosine (NPY) and two of its receptors (Y1- and Y2Rs) in different types of rodent dorsal root ganglion (DRG) and spinal cord neurons, and their regulation by peripheral nerve injury, have suggested a role in neuropathic pain. Here we present the spinal NPYergic system from an immunohistochemical perspective based on recent studies using two specific antibodies recognizing the Y1- and Y2Rs, respectively, as well as on data from a study on a Y1R knock-out mouse. We have, for example, defined seven different neuron populations of Y1R-expressing neurons in the rat spinal cord, representing multiple targets for spinally released NPY. The differential distribution of NPY receptors probably explains both the pro- and antinociceptive effects of NPY previously reported in the literature. One system possibly responsible for antinociception is a group of Y1R-positive, presumably glutamatergic interneurons in the superficial dorsal horn laminae. We also discuss the possibility that NPY released within DRGs can act in a paracrine fashion on NPY receptors on adjacent neurons, perhaps contributing to the so-called cross excitation, a concept advanced by Devor, Amir and collaborators. Taken together with behavioral and electrophysiological results summarized by Smith et al. in this volume, histochemical analyses have advanced the knowledge on the role of NPY in pain processing.
Collapse
Affiliation(s)
- Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
50
|
Smith PA, Moran TD, Abdulla F, Tumber KK, Taylor BK. Spinal mechanisms of NPY analgesia. Peptides 2007; 28:464-74. [PMID: 17194506 DOI: 10.1016/j.peptides.2006.09.029] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2006] [Accepted: 09/18/2006] [Indexed: 12/25/2022]
Abstract
We review previously published data, and present some new data, indicating that spinal application of neuropeptide Y (NPY) reduces behavioral and neurophysiological signs of acute and chronic pain. In models of acute pain, early behavioral studies showed that spinal (intrathecal) administration of NPY and Y2 receptor agonists decrease thermal nociception. Subsequent neurophysiological studies indicated that Y2-mediated inhibition of excitatory neurotransmitter release from primary afferent terminals in the substantia gelatinosa may contribute to the antinociceptive actions of NPY. As with acute pain, NPY reduced behavioral signs of inflammatory pain such as mechanical allodynia and thermal hyperalgesia; however, receptor antagonist studies indicate an important contribution of spinal Y1 rather than Y2 receptors. Interestingly, Y1 agonists suppress inhibitory synaptic events in dorsal horn neurons (indeed, well known mu-opioid analgesic drugs produce similar cellular actions). To resolve the behavioral and neurophysiological data, we propose that NPY/Y1 inhibits the spinal release of inhibitory neurotransmitters (GABA and glycine) onto inhibitory neurons, e.g. disinhibition of pain inhibition, resulting in hyporeflexia. The above mechanisms of Y1- and Y2-mediated analgesia may also operate in the setting of peripheral nerve injury, and new data indicate that NPY dose-dependently inhibits behavioral signs of neuropathic pain. Indeed, neurophysiological studies indicate that Y2-mediated inhibition of Ca(2+) channel currents in dorsal root ganglion neurons is actually increased after axotomy. We conclude that spinal delivery of Y1 agonists may be of use in the treatment of chronic inflammatory pain, and that the use of Y1 and Y2 agonists in neuropathic pain warrants further consideration.
Collapse
Affiliation(s)
- Peter A Smith
- Department of Pharmacology and Centre for Neuroscience, University of Alberta, 9.75 Medical Sciences Building, Edmonton, Alberta T6G 2H7, Canada.
| | | | | | | | | |
Collapse
|