1
|
Rodriguez FD, Covenas R. Association of Neurokinin-1 Receptor Signaling Pathways with Cancer. Curr Med Chem 2024; 31:6460-6486. [PMID: 37594106 DOI: 10.2174/0929867331666230818110812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 06/14/2023] [Accepted: 07/01/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND Numerous biochemical reactions leading to altered cell proliferation cause tumorigenesis and cancer treatment resistance. The mechanisms implicated include genetic and epigenetic changes, modified intracellular signaling, and failure of control mechanisms caused by intrinsic and extrinsic factors alone or combined. No unique biochemical events are responsible; entangled molecular reactions conduct the resident cells in a tissue to display uncontrolled growth and abnormal migration. Copious experimental research supports the etiological responsibility of NK-1R (neurokinin-1 receptor) activation, alone or cooperating with other mechanisms, in cancer appearance in different tissues. Consequently, a profound study of this receptor system in the context of malignant processes is essential to design new treatments targeting NK-1R-deviated activity. METHODS This study reviews and discusses recent literature that analyzes the main signaling pathways influenced by the activation of neurokinin 1 full and truncated receptor variants. Also, the involvement of NK-1R in cancer development is discussed. CONCLUSION NK-1R can signal through numerous pathways and cross-talk with other receptor systems. The participation of override or malfunctioning NK-1R in malignant processes needs a more precise definition in different types of cancers to apply satisfactory and effective treatments. A long way has already been traveled: the current disposal of selective and effective NK-1R antagonists and the capacity to develop new drugs with biased agonistic properties based on the receptor's structural states with functional significance opens immediate research action and clinical application.
Collapse
Affiliation(s)
- Francisco David Rodriguez
- Department of Biochemistry and Molecular Biology, Faculty of Chemical Sciences, University of Salamanca, 37007 Salamanca, Spain
- Group GIR USAL: BMD (Bases Moleculares del Desarrollo), University of Salamanca, Salamanca, Spain
| | - Rafael Covenas
- Group GIR USAL: BMD (Bases Moleculares del Desarrollo), University of Salamanca, Salamanca, Spain
- Laboratory of Neuroanatomy of the Peptidergic Systems, Institute of Neurosciences of Castilla y León (INCYL), University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
2
|
Babwah AV. The wonderful and masterful G protein-coupled receptor (GPCR): A focus on signaling mechanisms and the neuroendocrine control of fertility. Mol Cell Endocrinol 2020; 515:110886. [PMID: 32574585 DOI: 10.1016/j.mce.2020.110886] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/08/2020] [Accepted: 05/20/2020] [Indexed: 12/18/2022]
Abstract
Human GnRH deficiency, both clinically and genetically, is a heterogeneous disorder comprising of congenital GnRH deficiency with anosmia (Kallmann syndrome), or with normal olfaction [normosmic idiopathic hypogonadotropic hypogonadism (IHH)], and adult-onset hypogonadotropic hypogonadism. Our understanding of the neural mechanisms underlying GnRH secretion and GnRH signaling continues to increase at a rapid rate and strikingly, the heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors (GPCRs) continue to emerge as essential players in these processes. GPCRs were once viewed as binary on-off switches, where in the "on" state they are bound to their Gα protein, but now we understand that view is overly simplistic and does not adequately characterize GPCRs. Instead, GPCRs have emerged as masterful signaling molecules exploiting different physical conformational states of itself to elicit an array of downstream signaling events via their G proteins and the β-arrestins. The "one receptor-multiple signaling conformations" model is likely an evolved strategy that can be used to our advantage as researchers have shown that targeting specific receptor conformations via biased ligands is proving to be a powerful tool in the effective treatment of human diseases. Can biased ligands be used to selectively modulate signaling by GPCR regulators of the neuroendocrine axis in the treatment of IHH? As discussed in this review, the grand possibility exists. However, while we are still very far from developing these treatments, this exciting likelihood can happen through a much greater mechanistic understanding of how GPCRs signal within the cell.
Collapse
Affiliation(s)
- Andy V Babwah
- Department of Pediatrics, Laboratory of Human Growth and Reproductive Development, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States; Child Health Institute of New Jersey, New Brunswick, NJ, United States.
| |
Collapse
|
3
|
Gurevich VV, Gurevich EV. The structural basis of the arrestin binding to GPCRs. Mol Cell Endocrinol 2019; 484:34-41. [PMID: 30703488 PMCID: PMC6377262 DOI: 10.1016/j.mce.2019.01.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/04/2019] [Accepted: 01/17/2019] [Indexed: 12/12/2022]
Abstract
G protein-coupled receptors (GPCRs) are the largest family of signaling proteins targeted by more clinically used drugs than any other protein family. GPCR signaling via G proteins is quenched (desensitized) by the phosphorylation of the active receptor by specific GPCR kinases (GRKs) followed by tight binding of arrestins to active phosphorylated receptors. Thus, arrestins engage two types of receptor elements: those that contain GRK-added phosphates and those that change conformation upon activation. GRKs attach phosphates to serines and threonines in the GPCR C-terminus or any one of the cytoplasmic loops. In addition to these phosphates, arrestins engage the cavity that appears between trans-membrane helices upon receptor activation and several other non-phosphorylated elements. The residues that bind GPCRs are localized on the concave side of both arrestin domains. Arrestins undergo a global conformational change upon receptor binding (become activated). Arrestins serve as important hubs of cellular signaling, emanating from activated GPCRs and receptor-independent.
Collapse
Affiliation(s)
- Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.
| | - Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
| |
Collapse
|
4
|
Spitsin S, Pappa V, Douglas SD. Truncation of neurokinin-1 receptor-Negative regulation of substance P signaling. J Leukoc Biol 2018; 103:1043-1051. [PMID: 29345372 DOI: 10.1002/jlb.3mir0817-348r] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/22/2017] [Accepted: 12/07/2017] [Indexed: 12/21/2022] Open
Abstract
Substance P (SP) is a tachykinin peptide, which triggers intracellular signaling in the nervous and immune systems, as well as, other local and systemic events. The interaction between SP and its receptor, neurokinin-1 receptor (NK1R), results in major downstream cellular actions, which include changes in calcium fluxes, ERK, and p21-activated kinase phosphorylation and NFκB activation. Two naturally occurring variants of the NK1R, the full-length, 407 aa receptor (NK1R-F) and the truncated, 311 aa isoform (NK1R-T), mediate the actions of SP. Receptor truncation partially disrupts signaling motifs of the carboxyl tail, a critical site for mediating NK1R signaling, resulting in a "less-efficient" receptor. Although NK1R-F is the predominant isoform in the central and peripheral nervous systems, NK1R-T is expressed in several tissues and cells, which include monocytes, NK cells, and T-cells. The SP binding domain is not affected by truncation and this site is identical in both NK1R receptor isoforms. However, while cells expressing NK1R-F respond to nanomolar concentrations of SP, monocyte and macrophage activation, mediated through NK1R-T, requires micromolar concentrations of SP in order to elicit signaling responses. Elevated plasma levels of SP are associated with increased inflammatory responses and NK1R antagonists reduce inflammation and cytokine production in vivo. This mini review presents and discusses the novel hypothesis that the expression of NK1R-T on immune system cells prevents immune activation in a milieu, which usually contains low concentrations of SP and, thus, maintains immune homeostasis. In contrast, in the activated neuronal microenvironment, when SP levels reach the threshold at tissue sites, SP promotes immune activation and modulates monocyte/macrophage polarization.
Collapse
Affiliation(s)
- Sergei Spitsin
- Department of Pediatrics, Division of Allergy and Immunology, Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
| | - Vasiliki Pappa
- Department of Pediatrics, Division of Allergy and Immunology, Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
| | - Steven D Douglas
- Department of Pediatrics, Division of Allergy and Immunology, Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
5
|
Reiter E, Ayoub MA, Pellissier LP, Landomiel F, Musnier A, Tréfier A, Gandia J, De Pascali F, Tahir S, Yvinec R, Bruneau G, Poupon A, Crépieux P. β-arrestin signalling and bias in hormone-responsive GPCRs. Mol Cell Endocrinol 2017; 449:28-41. [PMID: 28174117 DOI: 10.1016/j.mce.2017.01.052] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Revised: 01/31/2017] [Accepted: 01/31/2017] [Indexed: 12/14/2022]
Abstract
G protein-coupled receptors (GPCRs) play crucial roles in the ability of target organs to respond to hormonal cues. GPCRs' activation mechanisms have long been considered as a two-state process connecting the agonist-bound receptor to heterotrimeric G proteins. This view is now challenged as mounting evidence point to GPCRs being connected to large arrays of transduction mechanisms involving heterotrimeric G proteins as well as other players. Amongst the G protein-independent transduction mechanisms, those elicited by β-arrestins upon their recruitment to the active receptors are by far the best characterized and apply to most GPCRs. These concepts, in conjunction with remarkable advances made in the field of GPCR structural biology and biophysics, have supported the notion of ligand-selective signalling also known as pharmacological bias. Interestingly, recent reports have opened intriguing prospects to the way β-arrestins control GPCR-mediated signalling in space and time within the cells. In the present paper, we review the existing evidence linking endocrine-related GPCRs to β-arrestin recruitement, signalling, pathophysiological implications and selective activation by biased ligands and/or receptor modifications. Emerging concepts surrounding β-arrestin-mediated transduction are discussed in the light of the peculiarities of endocrine systems.
Collapse
Affiliation(s)
- Eric Reiter
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France.
| | - Mohammed Akli Ayoub
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France; LE STUDIUM(®) Loire Valley Institute for Advanced Studies, 45000, Orléans, France; Biology Department, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | | | - Flavie Landomiel
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Astrid Musnier
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Aurélie Tréfier
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Jorge Gandia
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | | | - Shifa Tahir
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Romain Yvinec
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Gilles Bruneau
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Anne Poupon
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Pascale Crépieux
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| |
Collapse
|
6
|
Shahzad R, Jones MR, Viles JH, Jones CE. Endocytosis of the tachykinin neuropeptide, neurokinin B, in astrocytes and its role in cellular copper uptake. J Inorg Biochem 2016; 162:319-325. [PMID: 26948444 DOI: 10.1016/j.jinorgbio.2016.02.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/27/2016] [Accepted: 02/25/2016] [Indexed: 01/06/2023]
Abstract
The tachykinin neuropeptide, neurokinin B (NKB), belongs to a family of peptides having diverse roles in the brain. NKB, along with several other tachykinins, has been identified as a copper-binding peptide, however the physiological relevance of the binding is unclear. Previously, NKB was shown to limit the ability of copper to enter astrocytes and disrupt calcium homeostasis and it was thought that the peptide was sequestering the metal extracellularly. Here we use a fluorescein-labelled NKB peptide (F-NKB) to show that NKB is not retained extracellularly, but is endocytosed within 10-20min after addition to the cell media. The endocytosis is not inhibited when NKB is delivered as a copper-complex, [CuII(F-NKB)2]. Endocytosis of NKB can increase intracellular copper. Comparison to cells cultured in copper-free buffer indicated that apo-NKB can facilitate uptake of copper found in normal culture media. To achieve this NKB must compete with a variety of copper proteins, and we show that NKB can successfully compete with copper-binding peptides derived from the prion protein, itself associated with Cu(II) and Zn(II) metabolism. We suggest a mechanism of receptor mediated endocytosis to account for the observations.
Collapse
Affiliation(s)
- Reeha Shahzad
- The School of Science and Health, Western Sydney University, Locked Bag 1797, Penrith, 2759, NSW, Australia
| | - Mark R Jones
- The School of Science and Health, Western Sydney University, Locked Bag 1797, Penrith, 2759, NSW, Australia
| | - John H Viles
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Christopher E Jones
- The School of Science and Health, Western Sydney University, Locked Bag 1797, Penrith, 2759, NSW, Australia.
| |
Collapse
|
7
|
Geppetti P, Veldhuis N, Lieu T, Bunnett N. G Protein-Coupled Receptors: Dynamic Machines for Signaling Pain and Itch. Neuron 2015; 88:635-49. [DOI: 10.1016/j.neuron.2015.11.001] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
8
|
Ahow M, Min L, Pampillo M, Nash C, Wen J, Soltis K, Carroll RS, Glidewell-Kenney CA, Mellon PL, Bhattacharya M, Tobet SA, Kaiser UB, Babwah AV. KISS1R signals independently of Gαq/11 and triggers LH secretion via the β-arrestin pathway in the male mouse. Endocrinology 2014; 155:4433-46. [PMID: 25147978 PMCID: PMC4197989 DOI: 10.1210/en.2014-1304] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hypothalamic GnRH is the master regulator of the neuroendocrine reproductive axis, and its secretion is regulated by many factors. Among these is kisspeptin (Kp), a potent trigger of GnRH secretion. Kp signals via the Kp receptor (KISS1R), a Gαq/11-coupled 7-transmembrane-spanning receptor. Until this study, it was understood that KISS1R mediates GnRH secretion via the Gαq/11-coupled pathway in an ERK1/2-dependent manner. We recently demonstrated that KISS1R also signals independently of Gαq/11 via β-arrestin and that this pathway also mediates ERK1/2 activation. Because GnRH secretion is ERK1/2-dependent, we hypothesized that KISS1R regulates GnRH secretion via both the Gαq/11- and β-arrestin-coupled pathways. To test this hypothesis, we measured LH secretion, a surrogate marker of GnRH secretion, in mice lacking either β-arrestin-1 or β-arrestin-2. Results revealed that Kp-dependent LH secretion was significantly diminished relative to wild-type mice (P < .001), thus supporting that β-arrestin mediates Kp-induced GnRH secretion. Based on this, we hypothesized that Gαq/11-uncoupled KISS1R mutants, like L148S, will display Gαq/11-independent signaling. To test this hypothesis, L148S was expressed in HEK 293 cells. and results confirmed that, although strongly uncoupled from Gαq/11, L148S retained the ability to trigger significant Kp-dependent ERK1/2 phosphorylation (P < .05). Furthermore, using mouse embryonic fibroblasts lacking β-arrestin-1 and -2, we demonstrated that L148S-mediated ERK1/2 phosphorylation is β-arrestin-dependent. Overall, we conclude that KISS1R signals via Gαq/11 and β-arrestin to regulate GnRH secretion. This novel and important finding could explain why patients bearing some types of Gαq/11-uncoupled KISS1R mutants display partial gonadotropic deficiency and even a reversal of the condition, idiopathic hypogonadotropic hypogonadism.
Collapse
|
9
|
Glidewell-Kenney CA, Trang C, Shao PP, Gutierrez-Reed N, Uzo-Okereke AM, Coss D, Mellon PL. Neurokinin B induces c-fos transcription via protein kinase C and activation of serum response factor and Elk-1 in immortalized GnRH neurons. Endocrinology 2014; 155:3909-19. [PMID: 25057795 PMCID: PMC4164922 DOI: 10.1210/en.2014-1263] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Mutations in neurokinin B (NKB) and its receptor, NK3R, were identified in human patients with hypogonadotropic hypogonadism, a disorder characterized by lack of puberty and infertility. Further studies have suggested that NKB acts at the level of the hypothalamus to control GnRH neuron activity, either directly or indirectly. We recently reported that treatment with senktide, a NK3R agonist, induced GnRH secretion and expression of c-fos mRNA in GT1-7 cells. Here, we map the responsive region in the murine c-fos promoter to between -400 and -200 bp, identify the signal transducer and activator of transcription (STAT) (-345) and serum response element (-310) sites as required for induction, a modulatory role for the Ets site (-318), and show that induction is protein kinase C dependent. Using gel shift and Gal4 assays, we further show that phosphorylation of Elk-1 leads to binding to DNA in complex with serum response factor at serum response element and Ets sites within the c-fos promoter. Thus, we determine molecular mechanisms involved in NKB regulation of c-fos induction, which may play a role in modulation of GnRH neuron activation.
Collapse
Affiliation(s)
- Christine A Glidewell-Kenney
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093-0674
| | | | | | | | | | | | | |
Collapse
|
10
|
Raehal KM, Bohn LM. β-arrestins: regulatory role and therapeutic potential in opioid and cannabinoid receptor-mediated analgesia. Handb Exp Pharmacol 2014; 219:427-43. [PMID: 24292843 PMCID: PMC4804701 DOI: 10.1007/978-3-642-41199-1_22] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pain is a complex disorder with neurochemical and psychological components contributing to the severity, the persistence, and the difficulty in adequately treating the condition. Opioid and cannabinoids are two classes of analgesics that have been used to treat pain for centuries and are arguably the oldest of "pharmacological" interventions used by man. Unfortunately, they also produce several adverse side effects that can complicate pain management. Opioids and cannabinoids act at G protein-coupled receptors (GPCRs), and much of their effects are mediated by the mu-opioid receptor (MOR) and cannabinoid CB1 receptor (CB1R), respectively. These receptors couple to intracellular second messengers and regulatory proteins to impart their biological effects. In this chapter, we review the role of the intracellular regulatory proteins, β-arrestins, in modulating MOR and CB1R and how they influence the analgesic and side-effect profiles of opioid and cannabinoid drugs in vivo. This review of the literature suggests that the development of opioid and cannabinoid agonists that bias MOR and CB1R toward G protein signaling cascades and away from β-arrestin interactions may provide a novel mechanism by which to produce analgesia with less severe adverse effects.
Collapse
MESH Headings
- Analgesics/adverse effects
- Analgesics/pharmacology
- Analgesics, Opioid/adverse effects
- Analgesics, Opioid/pharmacology
- Animals
- Arrestins/metabolism
- Cannabinoids/adverse effects
- Cannabinoids/pharmacology
- Drug Design
- Humans
- Pain/drug therapy
- Pain/physiopathology
- Receptor, Cannabinoid, CB1/drug effects
- Receptor, Cannabinoid, CB1/metabolism
- Receptors, G-Protein-Coupled/drug effects
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/metabolism
- beta-Arrestins
Collapse
Affiliation(s)
- Kirsten M Raehal
- The Scripps Research Institute, 130 Scripps Way #2A2, Jupiter, FL, 33458, USA,
| | | |
Collapse
|
11
|
Noel SD, Abreu AP, Xu S, Muyide T, Gianetti E, Tusset C, Carroll J, Latronico AC, Seminara SB, Carroll RS, Kaiser UB. TACR3 mutations disrupt NK3R function through distinct mechanisms in GnRH-deficient patients. FASEB J 2013; 28:1924-37. [PMID: 24376026 DOI: 10.1096/fj.13-240630] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neurokinin B (NKB) and its G-protein-coupled receptor, NK3R, have been implicated in the neuroendocrine control of GnRH release; however, little is known about the structure-function relationship of this ligand-receptor pair. Moreover, loss-of-function NK3R mutations cause GnRH deficiency in humans. Using missense mutations in NK3R we previously identified in patients with GnRH deficiency, we demonstrate that Y256H and Y315C NK3R mutations in the fifth and sixth transmembrane domains (TM5 and TM6), resulted in reduced whole-cell (79.3±7.2%) or plasma membrane (67.3±7.3%) levels, respectively, compared with wild-type (WT) NK3R, with near complete loss of inositol phosphate (IP) signaling, implicating these domains in receptor trafficking, processing, and/or stability. We further demonstrate in a FRET-based assay that R295S NK3R, in the third intracellular loop (IL3), bound NKB but impaired dissociation of Gq-protein subunits from the receptor compared with WT NK3R, which showed a 10.0 ± 1.3% reduction in FRET ratios following ligand binding, indicating activation of Gq-protein signaling. Interestingly, R295S NK3R, identified in the heterozygous state in a GnRH-deficient patient, also interfered with dissociation of G proteins and IP signaling from wild-type NK3R, indicative of dominant-negative effects. Collectively, our data illustrate roles for TM5 and TM6 in NK3R trafficking and ligand binding and for IL3 in NK3R signaling.
Collapse
Affiliation(s)
- Sekoni D Noel
- 1Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, 221 Longwood Ave., Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Arresting inflammation: contributions of plasma membrane and endosomal signalling to neuropeptide-driven inflammatory disease. Biochem Soc Trans 2013; 41:137-43. [PMID: 23356273 DOI: 10.1042/bst20120343] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
GPCR (G-protein-coupled receptor) signalling at the plasma membrane is under tight control. In the case of neuropeptides such as SP (substance P), plasma membrane signalling is regulated by cell-surface endopeptidases (e.g. neprilysin) that degrade extracellular neuropeptides, and receptor interaction with β-arrestins, which uncouple receptors from heterotrimeric G-proteins and mediate receptor endocytosis. By recruiting GPCRs, kinases and phosphatases to endocytosed GPCRs, β-arrestins assemble signalosomes that can mediate a second wave of signalling by internalized receptors. Endosomal peptidases, such as ECE-1 (endothelin-converting enzyme-1), can degrade SP in acidified endosomes, which destabilizes signalosomes and allows receptors, freed from β-arrestins, to recycle and resensitize. By disassembling signalosomes, ECE-1 terminates β-arrestin-mediated endosomal signalling. These mechanisms have been studied in model cell systems, and the relative importance of plasma membrane and endosomal signalling to complex pathophysiological processes, such as inflammation, pain and proliferation, is unclear. However, deletion or inhibition of metalloendopeptidases that control neuropeptide signalling at the plasma membrane and in endosomes has marked effects on inflammation. Neprilysin deletion exacerbates inflammation because of diminished degradation of pro-inflammatory SP. Conversely, inhibition of ECE-1 attenuates inflammation by preventing receptor recycling/resensitization, which is required for sustained pro-inflammatory signals from the plasma membrane. β-Arrestin deletion also affects inflammation because of the involvement of β-arrestins in pro-inflammatory signalling and migration of inflammatory cells. Knowledge of GPCR signalling in specific subcellular locations provides insights into pathophysiological processes, and can provide new opportunities for therapy. Selective targeting of β-arrestin-mediated endosomal signalling or of mechanisms of receptor recycling/resensitization may offer more effective and selective treatments than global targeting of cell-surface signalling.
Collapse
|
13
|
Glidewell-Kenney CA, Shao PP, Iyer AK, Grove AMH, Meadows JD, Mellon PL. Neurokinin B causes acute GnRH secretion and repression of GnRH transcription in GT1-7 GnRH neurons. Mol Endocrinol 2013; 27:437-54. [PMID: 23393128 DOI: 10.1210/me.2012-1271] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Genetic studies in human patients with idiopathic hypogonadotropic hypogonadism (IHH) identified mutations in the genes that encode neurokinin B (NKB) and the neurokinin 3 receptor (NK3R). However, determining the mechanism whereby NKB regulates gonadotropin secretion has been difficult because of conflicting results from in vivo studies investigating the luteinizing hormone (LH) response to senktide, a NK3R agonist. NK3R is expressed in a subset of GnRH neurons and in kisspeptin neurons that are known to regulate GnRH secretion. Thus, one potential source of inconsistency is that NKB could produce opposing direct and indirect effects on GnRH secretion. Here, we employ the GT1-7 cell model to elucidate the direct effects of NKB on GnRH neuron function. We find that GT1-7 cells express NK3R and respond to acute senktide treatment with c-Fos induction and increased GnRH secretion. In contrast, long-term senktide treatment decreased GnRH secretion. Next, we focus on the examination of the mechanism underlying the long-term decrease in secretion and determine that senktide treatment represses transcription of GnRH. We further show that this repression of GnRH transcription may involve enhanced c-Fos protein binding at novel activator protein-1 (AP-1) half-sites identified in enhancer 1 and the promoter, as well as chromatin remodeling at the promoter of the GnRH gene. These data indicate that NKB could directly regulate secretion from NK3R-expressing GnRH neurons. Furthermore, whether the response is inhibitory or stimulatory toward GnRH secretion could depend on the history or length of exposure to NKB because of a repressive effect on GnRH transcription.
Collapse
Affiliation(s)
- Christine A Glidewell-Kenney
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093-0674, USA
| | | | | | | | | | | |
Collapse
|
14
|
Brioude F, Bouligand J, Francou B, Fagart J, Roussel R, Viengchareun S, Combettes L, Brailly-Tabard S, Lombès M, Young J, Guiochon-Mantel A. Two families with normosmic congenital hypogonadotropic hypogonadism and biallelic mutations in KISS1R (KISS1 receptor): clinical evaluation and molecular characterization of a novel mutation. PLoS One 2013; 8:e53896. [PMID: 23349759 PMCID: PMC3548821 DOI: 10.1371/journal.pone.0053896] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2012] [Accepted: 12/04/2012] [Indexed: 11/19/2022] Open
Abstract
Context KISS1R mutations have been reported in few patients with normosmic congenital hypogonadotropic hypogonadism (nCHH) (OMIM #146110). Objective To describe in detail nCHH patients with biallelic KISS1R mutations belonging to 2 unrelated families, and to functionally characterize a novel KISS1R mutation. Results An original mutant, p.Tyr313His, was found in the homozygous state in 3 affected kindred (2 females and 1 male) from a consanguineous Portuguese family. This mutation, located in the seventh transmembrane domain, affects a highly conserved amino acid, perturbs the conformation of the transmembrane segment, and impairs MAP kinase signaling and intracellular calcium release. In the second family, a French Caucasian male patient with nCHH was found to carry two recurrent mutations in the compound heterozygous state (p.Leu102Pro/Stop399Arg). In this man, pulsatile GnRH (Gonadotropin Releasing Hormone) administration restored pulsatile LH (Luteinizing Hormone) secretion and testicular hormone secretion. Later, long-term combined gonadotropin therapy induced spermatogenesis, enabling 3 successive pregnancies that resulted in 2 miscarriages and the birth of a healthy boy. Conclusion We show that a novel loss-of-function mutation (p.Tyr313His) in the KISS1R gene can cause familial nCHH, revealing the crucial role of this amino acid in KISS1R function. The observed restoration of gonadotropin secretion by exogenous GnRH administration further supports, in humans, the hypothalamic origin of the gonadotropin deficiency in this genetic form of nCHH.
Collapse
Affiliation(s)
- Frédéric Brioude
- Faculté de Médecine Paris-Sud UMR-S693, Univ Paris-Sud, Le Kremlin Bicêtre, France
- INSERM U693, IFR93, Le Kremlin Bicêtre, France
| | - Jérôme Bouligand
- Faculté de Médecine Paris-Sud UMR-S693, Univ Paris-Sud, Le Kremlin Bicêtre, France
- INSERM U693, IFR93, Le Kremlin Bicêtre, France
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - Bruno Francou
- Faculté de Médecine Paris-Sud UMR-S693, Univ Paris-Sud, Le Kremlin Bicêtre, France
- INSERM U693, IFR93, Le Kremlin Bicêtre, France
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - Jérôme Fagart
- Faculté de Médecine Paris-Sud UMR-S693, Univ Paris-Sud, Le Kremlin Bicêtre, France
- INSERM U693, IFR93, Le Kremlin Bicêtre, France
| | - Ronan Roussel
- Université Paris–Diderot, Paris 7, Paris, France
- Département d'Endocrinologie Diabétologie et Nutrition, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Say Viengchareun
- Faculté de Médecine Paris-Sud UMR-S693, Univ Paris-Sud, Le Kremlin Bicêtre, France
- INSERM U693, IFR93, Le Kremlin Bicêtre, France
| | - Laurent Combettes
- Faculté des Sciences, INSERM UMR-S757, Univ Paris-Sud, Orsay, France
| | - Sylvie Brailly-Tabard
- Faculté de Médecine Paris-Sud UMR-S693, Univ Paris-Sud, Le Kremlin Bicêtre, France
- INSERM U693, IFR93, Le Kremlin Bicêtre, France
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - Marc Lombès
- Faculté de Médecine Paris-Sud UMR-S693, Univ Paris-Sud, Le Kremlin Bicêtre, France
- INSERM U693, IFR93, Le Kremlin Bicêtre, France
- Service d'Endocrinologie et des Maladies de la Reproduction and Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jacques Young
- Faculté de Médecine Paris-Sud UMR-S693, Univ Paris-Sud, Le Kremlin Bicêtre, France
- INSERM U693, IFR93, Le Kremlin Bicêtre, France
- Service d'Endocrinologie et des Maladies de la Reproduction and Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris, Paris, France
- * E-mail: (JY); (AGM)
| | - Anne Guiochon-Mantel
- Faculté de Médecine Paris-Sud UMR-S693, Univ Paris-Sud, Le Kremlin Bicêtre, France
- INSERM U693, IFR93, Le Kremlin Bicêtre, France
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, France
- * E-mail: (JY); (AGM)
| |
Collapse
|
15
|
Orel M, Padrós E, Manyosa J. Structural features of the C-terminus from the human neurokinin-1 receptor. FEBS J 2012; 279:2357-67. [DOI: 10.1111/j.1742-4658.2012.08614.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
16
|
Ray P, Mihalko LA, Coggins NL, Moudgil P, Ehrlich A, Luker KE, Luker GD. Carboxy-terminus of CXCR7 regulates receptor localization and function. Int J Biochem Cell Biol 2012; 44:669-78. [PMID: 22300987 DOI: 10.1016/j.biocel.2012.01.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 01/10/2012] [Accepted: 01/16/2012] [Indexed: 01/09/2023]
Abstract
Chemokine receptor CXCR7 is essential for normal development, and this receptor promotes initiation and progression of diseases including cancer and autoimmunity. To understand normal and pathologic functions of CXCR7 and advance development of therapeutic agents, there is a need to define structural domains that regulate this receptor. We generated mutants of CXCR7 with deletion of different lengths of the predicted intracellular tail and analyzed effects on CXCR7 signaling and function in cell-based assays. While wild-type CXCR7 predominantly localized to intracellular vesicles, progressive deletion of the carboxy terminus redistributed the receptor to the plasma membrane. Truncating the intracellular tail of CXCR7 did not alter binding to CXCL12, but mutant receptors had reduced scavenging of this chemokine. Using a firefly luciferase complementation system, we established that deletions of the carboxy terminus decreased basal interactions and eliminated ligand-dependent recruitment of the scaffolding protein β-arrestin-2 to receptors. Deleting the carboxy terminus of CXCR7 impaired constitutive internalization of the receptor and reduced activation of ERK1/2 by CXCL12-CXCR7. Inhibiting dynamin, a molecule required for internalization of CXCR7, increased ligand-dependent association of the receptor with β-arrestin-2 and enhanced activation of ERK1/2. These studies establish mechanisms of action for CXCR7 and establish the intracellular tail of CXCR7 as a critical determinant of receptor trafficking, chemokine scavenging, and signaling.
Collapse
Affiliation(s)
- Paramita Ray
- Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Francou B, Bouligand J, Voican A, Amazit L, Trabado S, Fagart J, Meduri G, Brailly-Tabard S, Chanson P, Lecomte P, Guiochon-Mantel A, Young J. Normosmic congenital hypogonadotropic hypogonadism due to TAC3/TACR3 mutations: characterization of neuroendocrine phenotypes and novel mutations. PLoS One 2011; 6:e25614. [PMID: 22031817 PMCID: PMC3198730 DOI: 10.1371/journal.pone.0025614] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Accepted: 09/07/2011] [Indexed: 01/15/2023] Open
Abstract
CONTEXT TAC3/TACR3 mutations have been reported in normosmic congenital hypogonadotropic hypogonadism (nCHH) (OMIM #146110). In the absence of animal models, studies of human neuroendocrine phenotypes associated with neurokinin B and NK3R receptor dysfunction can help to decipher the pathophysiology of this signaling pathway. OBJECTIVE To evaluate the prevalence of TAC3/TACR3 mutations, characterize novel TACR3 mutations and to analyze neuroendocrine profiles in nCHH caused by deleterious TAC3/TACR3 biallelic mutations. RESULTS From a cohort of 352 CHH, we selected 173 nCHH patients and identified nine patients carrying TAC3 or TACR3 variants (5.2%). We describe here 7 of these TACR3 variants (1 frameshift and 2 nonsense deleterious mutations and 4 missense variants) found in 5 subjects. Modeling and functional studies of the latter demonstrated the deleterious consequence of one missense mutation (Tyr267Asn) probably caused by the misfolding of the mutated NK3R protein. We found a statistically significant (p<0.0001) higher mean FSH/LH ratio in 11 nCHH patients with TAC3/TACR3 biallelic mutations than in 47 nCHH patients with either biallelic mutations in KISS1R, GNRHR, or with no identified mutations and than in 50 Kallmann patients with mutations in KAL1, FGFR1 or PROK2/PROKR2. Three patients with TAC3/TACR3 biallelic mutations had an apulsatile LH profile but low-frequency alpha-subunit pulses. Pulsatile GnRH administration increased alpha-subunit pulsatile frequency and reduced the FSH/LH ratio. CONCLUSION The gonadotropin axis dysfunction associated with nCHH due to TAC3/TACR3 mutations is related to a low GnRH pulsatile frequency leading to a low frequency of alpha-subunit pulses and to an elevated FSH/LH ratio. This ratio might be useful for pre-screening nCHH patients for TAC3/TACR3 mutations.
Collapse
Affiliation(s)
- Bruno Francou
- Univ Paris-Sud, Faculté de Médecine Paris-Sud UMR-S693, Le Kremlin Bicêtre, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Le Kremlin Bicêtre, France
- INSERM U693, IFR93, Le Kremlin-Bicêtre, France
| | - Jérôme Bouligand
- Univ Paris-Sud, Faculté de Médecine Paris-Sud UMR-S693, Le Kremlin Bicêtre, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Le Kremlin Bicêtre, France
- INSERM U693, IFR93, Le Kremlin-Bicêtre, France
| | - Adela Voican
- Univ Paris-Sud, Faculté de Médecine Paris-Sud UMR-S693, Le Kremlin Bicêtre, France
- INSERM U693, IFR93, Le Kremlin-Bicêtre, France
- Universitatea de medicina si farmacie, Craiova, Romania
| | - Larbi Amazit
- Univ Paris-Sud, Faculté de Médecine Paris-Sud UMR-S693, Le Kremlin Bicêtre, France
- INSERM U693, IFR93, Le Kremlin-Bicêtre, France
| | - Séverine Trabado
- Univ Paris-Sud, Faculté de Médecine Paris-Sud UMR-S693, Le Kremlin Bicêtre, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Le Kremlin Bicêtre, France
- INSERM U693, IFR93, Le Kremlin-Bicêtre, France
| | | | - Geri Meduri
- Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Le Kremlin Bicêtre, France
- INSERM U693, IFR93, Le Kremlin-Bicêtre, France
| | - Sylvie Brailly-Tabard
- Univ Paris-Sud, Faculté de Médecine Paris-Sud UMR-S693, Le Kremlin Bicêtre, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Le Kremlin Bicêtre, France
- INSERM U693, IFR93, Le Kremlin-Bicêtre, France
| | - Philippe Chanson
- Univ Paris-Sud, Faculté de Médecine Paris-Sud UMR-S693, Le Kremlin Bicêtre, France
- INSERM U693, IFR93, Le Kremlin-Bicêtre, France
- Service d'Endocrinologie et des Maladies de la Reproduction and Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Le Kremlin Bicêtre, France
| | - Pierre Lecomte
- Service d'Endocrinologie, Hôpital Bretonneau, Tours, France
| | - Anne Guiochon-Mantel
- Univ Paris-Sud, Faculté de Médecine Paris-Sud UMR-S693, Le Kremlin Bicêtre, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Le Kremlin Bicêtre, France
- INSERM U693, IFR93, Le Kremlin-Bicêtre, France
| | - Jacques Young
- Univ Paris-Sud, Faculté de Médecine Paris-Sud UMR-S693, Le Kremlin Bicêtre, France
- INSERM U693, IFR93, Le Kremlin-Bicêtre, France
- Service d'Endocrinologie et des Maladies de la Reproduction and Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Le Kremlin Bicêtre, France
| |
Collapse
|
18
|
Murphy JE, Roosterman D, Cottrell GS, Padilla BE, Feld M, Brand E, Cedron WJ, Bunnett NW, Steinhoff M. Protein phosphatase 2A mediates resensitization of the neurokinin 1 receptor. Am J Physiol Cell Physiol 2011; 301:C780-91. [PMID: 21795521 DOI: 10.1152/ajpcell.00096.2011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Activated G protein-coupled receptors (GPCRs) are phosphorylated and interact with β-arrestins, which mediate desensitization and endocytosis. Endothelin-converting enzyme-1 (ECE-1) degrades neuropeptides in endosomes and can promote recycling. Although endocytosis, dephosphorylation, and recycling are accepted mechanisms of receptor resensitization, a large proportion of desensitized receptors can remain at the cell surface. We investigated whether reactivation of noninternalized, desensitized (phosphorylated) receptors mediates resensitization of the substance P (SP) neurokinin 1 receptor (NK(1)R). Herein, we report a novel mechanism of resensitization by which protein phosphatase 2A (PP2A) is recruited to dephosphorylate noninternalized NK(1)R. A desensitizing concentration of SP reduced cell-surface SP binding sites by only 25%, and SP-induced Ca(2+) signals were fully resensitized before cell-surface binding sites started to recover, suggesting resensitization of cell-surface-retained NK(1)R. SP induced association of β-arrestin1 and PP2A with noninternalized NK(1)R. β-Arrestin1 small interfering RNA knockdown prevented SP-induced association of cell-surface NK(1)R with PP2A, indicating that β-arrestin1 mediates this interaction. ECE-1 inhibition, by trapping β-arrestin1 in endosomes, also impeded SP-induced association of cell-surface NK(1)R with PP2A. Resensitization of NK(1)R signaling required both PP2A and ECE-1 activity. Thus, after stimulation with SP, PP2A interacts with noninternalized NK(1)R and mediates resensitization. PP2A interaction with NK(1)R requires β-arrestin1. ECE-1 promotes this process by releasing β-arrestin1 from NK(1)R in endosomes. These findings represent a novel mechanism of PP2A- and ECE-1-dependent resensitization of GPCRs.
Collapse
Affiliation(s)
- Jane E Murphy
- Department of Surgery, University of California, San Francisco, 94143-0660, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Malherbe P, Knoflach F, Hernandez MC, Hoffmann T, Schnider P, Porter RH, Wettstein JG, Ballard TM, Spooren W, Steward L. Characterization of RO4583298 as a novel potent, dual antagonist with in vivo activity at tachykinin NK₁ and NK₃ receptors. Br J Pharmacol 2011; 162:929-46. [PMID: 21039418 DOI: 10.1111/j.1476-5381.2010.01096.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Clinical results of osanetant and talnetant (selective-NK₃ antagonists) indicate that blocking the NK₃ receptor could be beneficial for the treatment of schizophrenia. The objective of this study was to characterize the in vitro and in vivo properties of a novel dual NK₁/NK₃ antagonist, RO4583298 (2-phenyl-N-(pyridin-3-yl)-N-methylisobutyramide derivative). EXPERIMENTAL APPROACH RO4583298 in vitro pharmacology was investigated using radioligand binding ([³H]-SP, [³H]-osanetant, [³H]-senktide), [³H]-inositol-phosphate accumulation Schild analysis (SP- or [MePhe⁷]-NKB-induced) and electrophysiological studies in guinea-pig substantia nigra pars compacta (SNpc). The in vivo activity of RO4583298 was assessed using reversal of GR73632-induced foot tapping in gerbils (GFT; NK₁) and senktide-induced tail whips in mice (MTW; NK₃). KEY RESULTS RO4583298 has a high-affinity for NK₁ (human and gerbil) and NK₃ (human, cynomolgus monkey, gerbil and guinea-pig) receptors and behaves as a pseudo-irreversible antagonist. Unusually it binds with high-affinity to mouse and rat NK₃, yet with a partial non-competitive mode of antagonism. In guinea-pig SNpc, RO4583298 inhibited the senktide-induced potentiation of spontaneous activity of dopaminergic neurones with an apparent non-competitive mechanism of action. RO4583298 (p.o.) robustly blocked the GFT response, and inhibited the MTW. CONCLUSIONS AND IMPLICATIONS RO4583298 is a high-affinity, non-competitive, long-acting in vivo NK₁/NK₃ antagonist; hence providing a useful in vitro and in vivo pharmacological tool to investigate the roles of NK₁ and NK₃ receptors in psychiatric disorders.
Collapse
Affiliation(s)
- P Malherbe
- Discovery Research CNS, F. Hoffmann-La Roche Ltd., Basel, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Monaco-Shawver L, Schwartz L, Tuluc F, Guo CJ, Lai JP, Gunnam SM, Kilpatrick LE, Banerjee PP, Douglas SD, Orange JS. Substance P inhibits natural killer cell cytotoxicity through the neurokinin-1 receptor. J Leukoc Biol 2010; 89:113-25. [PMID: 20940324 DOI: 10.1189/jlb.0410200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
SP is a potent neuroimmunomodulator that functions through ligating members of the neurokinin receptor family, one of which, NK1R, is widely expressed in immune cells. As in humans, circulating SP levels are increased in pathologic states associated with impairment of NK cell functions, such as depression and HIV infection, we hypothesized that SP has a direct, inhibitory effect upon NK cells. We have studied a clonal human NK cell line (YTS) as well as ex vivo human NK cells and have determined that truncated and full-length NK1R isoforms are expressed in and SP bound by ex vivo NK cells and the YTS NK cell line. Incubation of YTS cells with 10⁻⁶ M SP and ex vivo NK cells with 10⁻⁵ M SP inhibited cytotoxic ability by ∼20% and reduced degranulation. This inhibitory effect upon cytotoxicity was partially prevented by the NK1R antagonist CP96,345. The treatment of YTS or ex vivo NK cells with SP neither down-modulated NCR expression nor affected triggering receptor-induced NF-κB activation. Preincubation of YTS cells with SP, however, did abbreviate the typically prolonged intracellular calcium increase induced by target cell engagement and reduced triggering receptor-induced pERK. Thus, SP has the potential to regulate NK cell functions and acts downstream from neurokinin receptors to modulate NK cell activation signaling. This mechanism may contribute to impairment of NK cell function in certain disease states associated with increased circulating SP. Antagonism of this system may present an opportunity to augment NK cell function therapeutically in selected human diseases.
Collapse
Affiliation(s)
- Linda Monaco-Shawver
- The Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Pediatrics, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Vinet-Oliphant H, Alvarez X, Buza E, Borda JT, Mohan M, Aye PP, Tuluc F, Douglas SD, Lackner AA. Neurokinin-1 receptor (NK1-R) expression in the brains of SIV-infected rhesus macaques: implications for substance P in NK1-R immune cell trafficking into the CNS. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:1286-97. [PMID: 20671267 DOI: 10.2353/ajpath.2010.091109] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Recent studies suggest a link between neuropsychiatric disorders and HIV/SIV infection. Most evidence indicates that monocytes/macrophages are the primary cell type infected within the CNS and that they contribute to CNS inflammation and neurological disease. Substance P (SP), a pleotropic neuropeptide implicated in inflammation, depression, and immune modulation via interaction with its cognate receptor, the neurokinin 1 receptor (NK1-R), is produced by monocyte/macrophages. While the presence of NK1-R on neurons is well known, its role on cells of the immune system such as monocyte/macrophages is just beginning to emerge. Therefore, we have examined the expression of SP and NK1-R and their relationship to SIV/HIV encephalitis (SIVE/HIVE) lesions and SIV-infected cells. These studies demonstrated intense expression of SP and NK1-R in SIVE lesions, with macrophages being the principal cell expressing NK1-R. Interestingly, all of the SIV-infected macrophages expressed NK1-R. Additionally, we examined the functional role of SP as a proinflammatory mediator of monocyte activation and chemotaxis. These studies demonstrated that treatment of monocytes with SP elicited changes in cell-surface expression for CCR5 and NK1-R in a dose-dependent manner. Moreover, pretreatment with SP enhanced both SP- and CCL5-mediated chemotaxis. All of these findings suggest that SP and NK1-R are important in SIV infection of macrophages and the development of SIVE lesions.
Collapse
Affiliation(s)
- Heather Vinet-Oliphant
- Tulane National Primate Research Center, Division of Comparative Pathology, Covington, LA 70433, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Differences in the length of the carboxyl terminus mediate functional properties of neurokinin-1 receptor. Proc Natl Acad Sci U S A 2008; 105:12605-10. [PMID: 18713853 DOI: 10.1073/pnas.0806632105] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The neurokinin-1 receptor (NK1R) has two naturally occurring forms that differ in the length of the carboxyl terminus: a full-length receptor consisting of 407 aa and a truncated receptor consisting of 311 aa. We examined whether there are differential signaling properties attributable to the carboxyl terminus of this receptor by using stably transfected human embryonic kidney (HEK293) cell lines that express either full-length or truncated NK1R. Substance P (SP) specifically triggered intracellular calcium increase in HEK293 cells expressing full-length NK1R but had no effect in the cells expressing the truncated NK1R. In addition, in cells expressing full-length NK1R, SP activated NF-kappaB and IL-8 mRNA expression, but in cells expressing the truncated NK1R, SP did not activate NF-kappaB, and it decreased IL-8 mRNA expression. In cells expressing full-length NK1R, SP stimulated phosphorylation of PKCdelta but inhibited phosphorylation of PKCdelta in cells expressing truncated NK1R. There are also differences in the timing of SP-induced ERK activation in cells expressing the two different forms of the receptor. Full-length NK1R activation of ERK was rapid (peak within 1-2 min), whereas truncated NK1R-mediated activation was slower (peak at 20-30 min). Thus, the carboxyl terminus of NK1R is the structural basis for differences in the functional properties of the full-length and truncated NK1R. These differences may provide important information toward the design of new NK1R receptor antagonists.
Collapse
|
23
|
Le Brun I, Dufour A, Crest M, Szabó G, Erdelyi F, Baude A. Differential expression of Nk1 and NK3 neurokinin receptors in neurons of the nucleus tractus solitarius and the dorsal vagal motor nucleus of the rat and mouse. Neuroscience 2008; 152:56-64. [PMID: 18222044 DOI: 10.1016/j.neuroscience.2007.12.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Revised: 12/07/2007] [Accepted: 12/11/2007] [Indexed: 12/16/2022]
Abstract
Tachykinins (substance P, neurokinin A and neurokinin B) influence autonomic functions by modulating neuron activity in nucleus tractus solitarius (NTS) and dorsal motor nucleus of the vagus (DMV) through activation of neurokinin receptors NK1 and NK3. Our purpose was to identify and define by neurochemical markers, the subpopulations of NK1 and NK3 expressing neurons in NTS and DMV of rat and mouse. Because the distribution of the NK1 and NK3 expressing neurons overlaps, co-expression for both receptors was tested. By double labeling, we show that NK1 and NK3 were not co-expressed in NTS neurons. In the DMV, most of neurons (87%) were immunoreactive for only one of the receptors and 34% of NK1 neurons, 7% of NK3 neurons and 12% of NK1-NK3 neurons were cholinergic neurons. None of the neurons immunoreactive for NK1 or NK3 were positive for tyrosine hydroxylase, suggesting that catecholaminergic cells of the NTS (A2 and C2 groups) did not express neurokinin receptors. The presence of NK1 and NK3 was examined in GABAergic interneurons of the NTS and DMV by using GAD65-EGFP transgenic mouse. Immunoreactivity for NK1 or NK3 was found in a subpopulation of GAD65-EGFP cells. A majority (60%) of NK3 cells, but only 11% of the NK1 cells, were GAD65-EGFP cells. In conclusion, tachykinins, through differential expression of neurokinin receptors, may influence the central regulation of vital functions by acting on separate neuron subpopulations in NTS and DMV. Of particular interest, tachykinins may be involved in inhibitory mechanisms by acting directly on local GABAergic interneurons. Our results support a larger contribution of NK3 compared with NK1 in mediating inhibition in NTS and DMV.
Collapse
Affiliation(s)
- I Le Brun
- Laboratoire de Neurophysiologie Cellulaire, Université de la Méditerranée, CNRS UMR 6150, IFR Jean-Roche, Faculté de Médecine Nord, Boulevard Pierre Dramard, 13916 Marseille 20, France
| | | | | | | | | | | |
Collapse
|
24
|
Jensen D, Zhang Z, Flynn FW. Trafficking of tachykinin neurokinin 3 receptor to nuclei of neurons in the paraventricular nucleus of the hypothalamus following osmotic challenge. Neuroscience 2008; 155:308-16. [PMID: 18583062 DOI: 10.1016/j.neuroscience.2008.05.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2008] [Revised: 05/07/2008] [Accepted: 05/21/2008] [Indexed: 11/26/2022]
Abstract
Tachykinin neurokinin 3 receptor (NK3R) is a G-protein (GTP binding protein) -coupled receptor that is heavily expressed by magnocellular neurons of the paraventricular nucleus of the hypothalamus (PVN). Osmotic challenge is reported to activate NK3R expressed by magnocellular neurons and cause the NK3R to be internalized to the cytoplasm and perhaps the cell nucleus. In this study we show using immuno-electron microscopy that isolated nuclei from neurons in the PVN of osmotic challenged animals (rats) show a robust labeling for the NK3R. NK3R immunoreactivity was detected by Western blot in isolated nuclei of PVN neurons following the 2 M NaCl injection. No nuclear NK3R immunoreactivity was detected in control animals. NK3R antibody specificity was confirmed by small interfering (SI) RNA technology. This study establishes that the NK3R is trafficked to the nucleus of PVN neurons following a peripheral osmotic challenge.
Collapse
Affiliation(s)
- D Jensen
- Graduate Neuroscience Program, University of Wyoming, Department 3166, Laramie, WY 82071, USA
| | | | | |
Collapse
|
25
|
Poole DP, Amadesi S, Rozengurt E, Thacker M, Bunnett NW, Furness JB. Stimulation of the neurokinin 3 receptor activates protein kinase C epsilon and protein kinase D in enteric neurons. Am J Physiol Gastrointest Liver Physiol 2008; 294:G1245-56. [PMID: 18308856 DOI: 10.1152/ajpgi.00521.2007] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Tachykinins, acting through NK(3) receptors (NK(3)R), contribute to excitatory transmission to intrinsic primary afferent neurons (IPANs) of the small intestine. Although this transmission is dependent on protein kinase C (PKC), its maintenance could depend on protein kinase D (PKD), a downstream target of PKC. Here we show that PKD1/2-immunoreactivity occurred exclusively in IPANs of the guinea pig ileum, demonstrated by double staining with the IPAN marker NeuN. PKCepsilon was also colocalized with PKD1/2 in IPANs. PKCepsilon and PKD1/2 trafficking was studied in enteric neurons within whole mounts of the ileal wall. In untreated preparations, PKCepsilon and PKD1/2 were cytosolic and no signal for activated (phosphorylated) PKD was detected. The NK(3)R agonist senktide evoked a transient translocation of PKCepsilon and PKD1/2 from the cytosol to the plasma membrane and induced PKD1/2 phosphorylation at the plasma membrane. PKCepsilon translocation was maximal at 10 s and returned to the cytosol within 2 min. Phosphorylated-PKD1/2 was detected at the plasma membrane within 15 s and translocated to the cytosol by 2 min, where it remained active up to 30 min after NK(3)R stimulation. PKD1/2 activation was reduced by a PKCepsilon inhibitor and prevented by NK(3)R inhibition. NK(3)R-mediated PKCepsilon and PKD activation was confirmed in HEK293 cells transiently expressing NK(3)R and green fluorescent protein-tagged PKCepsilon, PKD1, PKD2, or PKD3. Senktide caused membrane translocation and activation of kinases within 30 s. After 15 min, phosphorylated PKD had returned to the cytosol. PKD activation was confirmed through Western blotting. Thus stimulation of NK(3)R activates PKCepsilon and PKD in sequence, and sequential activation of these kinases may account for rapid and prolonged modulation of IPAN function.
Collapse
Affiliation(s)
- D P Poole
- Department of Anatomy and Cell Biology and Centre for Neuroscience, University of Melbourne, Parkville, VIC, Australia.
| | | | | | | | | | | |
Collapse
|
26
|
Padilla BE, Cottrell GS, Roosterman D, Pikios S, Muller L, Steinhoff M, Bunnett NW. Endothelin-converting enzyme-1 regulates endosomal sorting of calcitonin receptor-like receptor and beta-arrestins. ACTA ACUST UNITED AC 2007; 179:981-97. [PMID: 18039931 PMCID: PMC2099187 DOI: 10.1083/jcb.200704053] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Although cell surface metalloendopeptidases degrade neuropeptides in the extracellular fluid to terminate signaling, the function of peptidases in endosomes is unclear. We report that isoforms of endothelin-converting enzyme-1 (ECE-1a–d) are present in early endosomes, where they degrade neuropeptides and regulate post-endocytic sorting of receptors. Calcitonin gene-related peptide (CGRP) co-internalizes with calcitonin receptor-like receptor (CLR), receptor activity-modifying protein 1 (RAMP1), β-arrestin2, and ECE-1 to early endosomes, where ECE-1 degrades CGRP. CGRP degradation promotes CLR/RAMP1 recycling and β-arrestin2 redistribution to the cytosol. ECE-1 inhibition or knockdown traps CLR/RAMP1 and β-arrestin2 in endosomes and inhibits CLR/RAMP1 recycling and resensitization, whereas ECE-1 overexpression has the opposite effect. ECE-1 does not regulate either the resensitization of receptors for peptides that are not ECE-1 substrates (e.g., angiotensin II), or the recycling of the bradykinin B2 receptor, which transiently interacts with β-arrestins. We propose a mechanism by which endosomal ECE-1 degrades neuropeptides in endosomes to disrupt the peptide/receptor/β-arrestin complex, freeing internalized receptors from β-arrestins and promoting recycling and resensitization.
Collapse
Affiliation(s)
- Benjamin E Padilla
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Roosterman D, Cottrell GS, Padilla BE, Muller L, Eckman CB, Bunnett NW, Steinhoff M. Endothelin-converting enzyme 1 degrades neuropeptides in endosomes to control receptor recycling. Proc Natl Acad Sci U S A 2007; 104:11838-43. [PMID: 17592116 PMCID: PMC1913888 DOI: 10.1073/pnas.0701910104] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Neuropeptide signaling requires the presence of G protein-coupled receptors (GPCRs) at the cell surface. Activated GPCRs interact with beta-arrestins, which mediate receptor desensitization, endocytosis, and mitogenic signaling, and the peptide-receptor-arrestin complex is sequestered into endosomes. Although dissociation of beta-arrestins is required for receptor recycling and resensitization, the critical event that initiates this process is unknown. Here we report that the agonist availability in the endosomes, controlled by the membrane metalloendopeptidase endothelin-converting enzyme 1 (ECE-1), determines stability of the peptide-receptor-arrestin complex and regulates receptor recycling and resensitization. Substance P (SP) binding to the tachykinin neurokinin 1 receptor (NK1R) induced membrane translocation of beta-arrestins followed by trafficking of the SP-NK1R-beta-arrestin complex to early endosomes containing ECE-1a-d. ECE-1 degraded SP in acidified endosomes, disrupting the complex; beta-arrestins returned to the cytosol, and the NK1R, freed from beta-arrestins, recycled and resensitized. An ECE-1 inhibitor, by preventing NK1R recycling in endothelial cells, inhibited resensitization of SP-induced inflammation. This mechanism is a general one because ECE-1 similarly regulated NK3R resensitization. Thus, peptide availability in endosomes, here regulated by ECE-1, determines the stability of the peptide-receptor-arrestin complex. This mechanism regulates receptor recycling, which is necessary for sustained signaling, and it may also control beta-arrestin-dependent mitogenic signaling of endocytosed receptors. We propose that other endosomal enzymes and transporters may similarly control the availability of transmitters in endosomes to regulate trafficking and signaling of GPCRs. Antagonism of these endosomal processes represents a strategy for inhibiting sustained signaling of receptors, and defects may explain the tachyphylaxis of drugs that are receptor agonists.
Collapse
Affiliation(s)
- Dirk Roosterman
- *Department of Dermatology, Interdisziplinäres Zentrum für Klinische Forschung Münster, and Ludwig Boltzmann Institute for Cell Biology and Immunobiology of the Skin, University of Münster, Von-Esmarch-Strasse 58, 48149 Münster, Germany
| | - Graeme S. Cottrell
- Departments of Surgery and Physiology, University of California, San Francisco, CA 94143
| | - Benjamin E. Padilla
- Departments of Surgery and Physiology, University of California, San Francisco, CA 94143
| | - Laurent Muller
- Institut National de la Santé et de la Recherche Médicale, Unité 36, College de France Paris, 75005 Paris, France; and
| | | | - Nigel W. Bunnett
- Departments of Surgery and Physiology, University of California, San Francisco, CA 94143
- To whom correspondence should be addressed at:
University of California, San Francisco, Room S1268, Box 0660, 513 Parnassus Avenue, San Francisco, CA 94143-0660. E-mail:
| | - Martin Steinhoff
- *Department of Dermatology, Interdisziplinäres Zentrum für Klinische Forschung Münster, and Ludwig Boltzmann Institute for Cell Biology and Immunobiology of the Skin, University of Münster, Von-Esmarch-Strasse 58, 48149 Münster, Germany
| |
Collapse
|
28
|
Ravina CG, Seda M, Pinto FM, Orea A, Fernández-Sánchez M, Pintado CO, Candenas ML. A role for tachykinins in the regulation of human sperm motility. Hum Reprod 2007; 22:1617-25. [PMID: 17437961 DOI: 10.1093/humrep/dem069] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Tachykinins and tachykinin receptors are widely distributed in the male reproductive tract and appear to be involved in reproduction. However, the function and expression of tachykinins and their receptors in human spermatozoa remain poorly studied. We analysed the effects of tachykinins on sperm motility and characterized the population of tachykinin receptors in human spermatozoa. METHODS AND RESULTS Motility analysis was performed following World Health Organization guidelines and we found that substance P (SP), human hemokinin-1 (hHK-1), neurokinin A (NKA) and neurokinin B (NKB) produced concentration-dependent increases in sperm progressive motility. The effects of tachykinins were antagonized by the NK(1) receptor-selective antagonist SR 140333, the NK(2) receptor-selective antagonist, SR 48968 and, to a lesser extent, also by the NK(3) receptor-selective antagonist SR 142801. Immunocytochemistry studies showed expression of the NK(1), NK(2) and NK(3) tachykinin receptor proteins in spermatozoa with different major sites of localization for each receptor. Western blot analysis confirmed the presence of tachykinin receptors in sperm cell homogenates. RT-PCR demonstrated expression of the genes that encode SP/NKA (TAC1), NKB (TAC3) and hHK-1 (TAC4) but not the genes TACR1, TACR2 and TACR3 encoding NK(1), NK(2) and NK(3) receptors, respectively. CONCLUSIONS These results show for the first time that the NK(1), NK(2) and NK(3) tachykinin receptor proteins are present in human spermatozoa. Our findings suggest that tachykinins, probably acting through these three tachykinin receptors, play a role in the regulation of human sperm motility.
Collapse
Affiliation(s)
- C G Ravina
- Instituto de Investigaciones Químicas, CSIC-Universidad de Sevilla, 49 Americo Vespucio Avenue, 41092 Seville, Spain
| | | | | | | | | | | | | |
Collapse
|
29
|
Silva E, Gomes P, Soares-da-Silva P. Overexpression of Na(+)/K (+)-ATPase parallels the increase in sodium transport and potassium recycling in an in vitro model of proximal tubule cellular ageing. J Membr Biol 2007; 212:163-75. [PMID: 17334838 DOI: 10.1007/s00232-005-7017-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2005] [Revised: 05/11/2006] [Indexed: 11/26/2022]
Abstract
Na(+)/K(+)-ATPase plays a key role in the transport of Na(+) throughout the nephron, but ageing appears to be accompanied by changes in the regulation and localization of the pump. In the present study, we examined the effect of in vitro cell ageing on the transport of Na(+) and K(+) ions in opossum kidney (OK) cells in culture. Cells were aged by repeated passing, and Na(+)/K(+)-ATPase activity and K(+) conductance were evaluated using electrophysiological methods. Na(+)K(+)-ATPase alpha(1)- and beta(1)-subunit expression was quantified by Western blot techniques. Na(+)/H(+) exchanger activity, changes in membrane potential, cell viability, hydrogen peroxide production and cellular proliferation were determined using fluorimetric assays. In vitro cell ageing is accompanied by an increase in transepithelial Na(+) transport, which results from an increase in the number of Na(+)/K(+)-ATPase alpha(1)- and beta(1)-subunits, in the membrane. Increases in Na(+)/K(+)-ATPase activity were accompanied by increases in K(+) conductance as a result of functional coupling between Na(+)/K(+)-ATPase and basolateral K(+) channels. Cell depolarization induced by both KCl and ouabain was more pronounced in aged cells. No changes in Na(+)/H(+) exchanger activity were observed. H(2)O(2) production was increased in aged cells, but exposure for 5 days to 1 and 10 microM: of H(2)O(2) had no effect on Na(+)/K(+)-ATPase expression. Ouabain (100 nM: ) increased alpha(1)-subunit, but not beta(1)-subunit, Na(+)/K(+)-ATPase expression in aged cells only. These cells constitute an interesting model for the study of renal epithelial cell ageing.
Collapse
Affiliation(s)
- E Silva
- Faculty of Medicine, Institute of Pharmacology and Therapeutics, 4200-319 , Porto, Portugal
| | | | | |
Collapse
|
30
|
Cottrell GS, Padilla B, Pikios S, Roosterman D, Steinhoff M, Grady EF, Bunnett NW. Post-endocytic sorting of calcitonin receptor-like receptor and receptor activity-modifying protein 1. J Biol Chem 2007; 282:12260-71. [PMID: 17310067 DOI: 10.1074/jbc.m606338200] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Calcitonin receptor-like receptor (CLR) and the receptor activity-modifying protein 1 (RAMP1) comprise a receptor for calcitonin gene-related peptide (CGRP). Although CGRP induces endocytosis of CLR/RAMP1, little is known about post-endocytic sorting of these proteins. We observed that the duration of stimulation with CGRP markedly affected post-endocytic sorting of CLR/RAMP1. In HEK and SK-N-MC cells, transient stimulation (10(-7) M CGRP, 1 h), induced CLR/RAMP1 recycling with similar kinetics (2-6 h), demonstrated by labeling receptors in living cells with antibodies to extracellular epitopes. Recycling of CLR/RAMP1 correlated with resensitization of CGRP-induced increases in [Ca(2+)](i). Cycloheximide did not affect resensitization, but bafilomycin A(1), an inhibitor of vacuolar H(+)-ATPases, abolished resensitization. Recycling CLR and RAMP1 were detected in endosomes containing Rab4a and Rab11a, and expression of GTPase-defective Rab4aS22N and Rab11aS25N inhibited resensitization. After sustained stimulation (10(-7) M CGRP, >2 h), CLR/RAMP1 trafficked to lysosomes. RAMP1 was degraded approximately 4-fold more rapidly than CLR (RAMP1, 45% degradation, 5 h; CLR, 54% degradation, 16 h), determined by Western blotting. Inhibitors of lysosomal, but not proteasomal, proteases prevented degradation. Sustained stimulation did not induce detectable mono- or polyubiquitination of CLR or RAMP1, determined by immunoprecipitation and Western blotting. Moreover, a RAMP1 mutant lacking the only intracellular lysine (RAMP1K142R) internalized and was degraded normally. Thus, after transient stimulation with CGRP, CLR and RAMP1 traffic from endosomes to the plasma membrane, which mediates resensitization. After sustained stimulation, CLR and RAMP1 traffic from endosomes to lysosomes by ubiquitin-independent mechanisms, where they are degraded at different rates.
Collapse
Affiliation(s)
- Graeme S Cottrell
- Department of Surgery, University of California, San Francisco, California 94143-0660, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Cottrell GS, Padilla B, Pikios S, Roosterman D, Steinhoff M, Gehringer D, Grady EF, Bunnett NW. Ubiquitin-dependent down-regulation of the neurokinin-1 receptor. J Biol Chem 2006; 281:27773-83. [PMID: 16849335 DOI: 10.1074/jbc.m603369200] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Transient stimulation with substance P (SP) induces endocytosis and recycling of the neurokinin-1 receptor (NK(1)R). The effects of sustained stimulation by high concentrations of SP on NK(1)R trafficking and Ca(2+) signaling, as may occur during chronic inflammation and pain, are unknown. Chronic exposure to SP (100 nm, 3 h) completely desensitized Ca(2+) signaling by wild-type NK(1)R (NK(1)Rwt). Resensitization occurred after 16 h, and cycloheximide prevented resensitization, implicating new receptor synthesis. Lysine ubiquitination of G-protein-coupled receptors is a signal for their trafficking and degradation. Lysine-deficient mutant receptors (NK(1)RDelta5K/R, C-terminal tail lysines; and NK(1)RDelta10K/R, all intracellular lysines) were expressed at the plasma membrane and were functional because they responded to SP by endocytosis and by mobilization of Ca(2+) ions. SP desensitized NK(1)Rwt, NK(1)RDelta5K/R, and NK(1)RDelta10K/R. However, NK(1)RDelta5K/R and NK(1)RDelta10K/R resensitized 4-8-fold faster than NK(1)Rwt by cycloheximide-independent mechanisms. NK(1)RDelta325 (a naturally occurring truncated variant) showed incomplete desensitization, followed by a marked sensitization of signaling. Upon labeling receptors in living cells using antibodies to extracellular epitopes, we observed that SP induced endocytosis of NK(1)Rwt, NK(1)RDelta5K/R, and NK(1)RDelta10K/R. After 4 h in SP-free medium, NK(1)RDelta5K/R and NK(1)RDelta10K/R recycled to the plasma membrane, whereas NK(1)Rwt remained internalized. SP induced ubiquitination of NK(1)Rwt and NK(1)RDelta5K/R as determined by immunoprecipitation under nondenaturing and denaturing conditions and detected with antibodies for mono- and polyubiquitin. NK(1)RDelta10K/R was not ubiquitinated. Whereas SP induced degradation of NK(1)Rwt, NK(1)RDelta5K/R and NK(1)RDelta10K/R showed approximately 50% diminished degradation. Thus, chronic stimulation with SP induces ubiquitination of the NK(1)R, which mediates its degradation and down-regulation.
Collapse
Affiliation(s)
- Graeme S Cottrell
- Departments of Surgery and Physiology, University of California, San Francisco, 94143-0660, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Gurevich VV, Gurevich EV. The structural basis of arrestin-mediated regulation of G-protein-coupled receptors. Pharmacol Ther 2006; 110:465-502. [PMID: 16460808 PMCID: PMC2562282 DOI: 10.1016/j.pharmthera.2005.09.008] [Citation(s) in RCA: 361] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2005] [Accepted: 09/22/2005] [Indexed: 12/23/2022]
Abstract
The 4 mammalian arrestins serve as almost universal regulators of the largest known family of signaling proteins, G-protein-coupled receptors (GPCRs). Arrestins terminate receptor interactions with G proteins, redirect the signaling to a variety of alternative pathways, and orchestrate receptor internalization and subsequent intracellular trafficking. The elucidation of the structural basis and fine molecular mechanisms of the arrestin-receptor interaction paved the way to the targeted manipulation of this interaction from both sides to produce very stable or extremely transient complexes that helped to understand the regulation of many biologically important processes initiated by active GPCRs. The elucidation of the structural basis of arrestin interactions with numerous non-receptor-binding partners is long overdue. It will allow the construction of fully functional arrestins in which the ability to interact with individual partners is specifically disrupted or enhanced by targeted mutagenesis. These "custom-designed" arrestin mutants will be valuable tools in defining the role of various interactions in the intricate interplay of multiple signaling pathways in the living cell. The identification of arrestin-binding sites for various signaling molecules will also set the stage for designing molecular tools for therapeutic intervention that may prove useful in numerous disorders associated with congenital or acquired disregulation of GPCR signaling.
Collapse
Affiliation(s)
- Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | | |
Collapse
|
33
|
Muto S, Tsuruoka S, Miyata Y, Fujimura A, Kusano E. Effect of trimethoprim-sulfamethoxazole on Na and K+ transport properties in the rabbit cortical collecting duct perfused in vitro. Nephron Clin Pract 2005; 102:p51-60. [PMID: 16286787 DOI: 10.1159/000089682] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2005] [Accepted: 07/20/2005] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND In this study, the membrane mechanisms of hyperkalemia caused by trimethoprim-sulfamethoxazole (TMP-SMX) combination antibiotics were assessed in the cortical collecting duct (CCD). METHODS We used the microelectrode technique and flux measurements, and examined the effects of TMP and SMX on electrical properties of the apical and basolateral membranes in the rabbit CCD perfused in vitro. RESULTS TMP in the lumen caused increases in apical membrane voltage, fractional apical membrane resistance (fRA), and transepithelial resistance (RT), all effects which were completely inhibited by luminal amiloride, but not by luminal Ba2+. The luminal TMP inhibited both net Na+ reabsorption and K+ secretion in the CCD. TMP in the bath slightly but significantly depolarized transepithelial voltage and basolateral membrane voltage without influencing fRA or RT. SMX in the lumen or bath had no effect on barrier voltages or resistances. CONCLUSION TMP mainly acts on the apical membrane of the CCD, inhibits the amiloride-sensitive macroscopic Na+ conductance in this membrane, and thereby decreases the net driving force for K+ exit across the membrane, resulting in an inhibition of K+ secretion. SMX in the lumen or bath had no effect on the CCD.
Collapse
Affiliation(s)
- Shigeaki Muto
- Department of Nephrology, Jichi Medical School, Minamikawachi, Kawachi, Tochigi, Japan.
| | | | | | | | | |
Collapse
|
34
|
Howe HE, Somponpun SJ, Sladek CD. Role of neurokinin 3 receptors in supraoptic vasopressin and oxytocin neurons. J Neurosci 2005; 24:10103-10. [PMID: 15537880 PMCID: PMC6730173 DOI: 10.1523/jneurosci.3164-04.2004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Neurokinin 3 receptors (NK3-Rs) are expressed in the supraoptic nucleus (SON), and SON is innervated by substance P (SP)-expressing A1 neurons in the medulla. Because SP stimulates vasopressin (VP) and oxytocin release from explants of the hypothalamo-neurohypophyseal system (HNS), two hypotheses were tested: (1) SP-stimulated VP release is mediated by NK3-Rs, and (2) stimulation of the A1 pathway by hypotension activates SON NK3-Rs. Senktide, an NK3-R agonist, stimulated VP release from HNS explants, but neither a neurokinin 1 receptor antagonist [L732,138 (N-acetyl-L-tryptophan 3,5-bis(tri-fluoromethyl)benzyl ester)] nor two NK3-R antagonists (SB222200 and SB235375) prevented SP-stimulated VP release. Because the affinity of these antagonists for rat NK-Rs may limit their efficacy, NK3-R internalization was used to assess the ability of SP to activate SON NK3-Rs. Senktide, SP, or vehicle was microinjected above SON. The brain was perfused 5 min after injection and stained for NK3-R immunoreactivity. Using confocal microscopy, the number of NK3-R-immunoreactive (-IR) endosomes was counted in a 5.6(2) mu region of cytoplasm in SON neurons. Senktide, but not SP or vehicle, significantly increased the number of NK3-R-IR endosomes in the cytoplasm. When hypotension was induced with hydralazine, NK3-R internalization was observed within 5 min (p < 0.005). A decrease in cytoplasmic NK3-R immunoreactivity was observed within 15 min of hypotension. Unexpectedly, both senktide and hypotension resulted in translocation of NK3-R-IR immunoreactivity to the nucleus. Thus, although these studies do not identify SP as the NK3-R ligand, they do provide evidence for hypotension-induced release of an endogenous tachykinin in SON and evidence suggesting a role for NK3-Rs in transcription regulation.
Collapse
MESH Headings
- Acetates/pharmacology
- Animals
- Catecholamines/physiology
- Cell Compartmentation
- Cell Nucleus/chemistry
- Cytoplasm/chemistry
- Endosomes/chemistry
- Hydralazine/pharmacology
- Hypotension/chemically induced
- Hypotension/physiopathology
- Hypothalamo-Hypophyseal System/physiopathology
- Hypothalamus, Anterior/metabolism
- Male
- Microinjections
- Microscopy, Confocal
- Neurons/drug effects
- Neurons/metabolism
- Neurons/ultrastructure
- Oxytocin/metabolism
- Peptide Fragments/pharmacology
- Quinolines/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptors, G-Protein-Coupled/drug effects
- Receptors, G-Protein-Coupled/physiology
- Receptors, Neurokinin-1/drug effects
- Receptors, Neurokinin-1/physiology
- Receptors, Neurokinin-3/drug effects
- Receptors, Neurokinin-3/physiology
- Substance P/analogs & derivatives
- Substance P/pharmacology
- Tachykinins/physiology
- Transcription, Genetic/drug effects
- Transcription, Genetic/physiology
- Tryptophan/analogs & derivatives
- Tryptophan/pharmacology
- Vasopressins/metabolism
Collapse
Affiliation(s)
- Heather E Howe
- Department of Physiology and Biophysics, University of Colorado Health Sciences Center, Denver, Colorado 80262, USA
| | | | | |
Collapse
|
35
|
Wei H, Ahn S, Barnes WG, Lefkowitz RJ. Stable Interaction between β-Arrestin 2 and Angiotensin Type 1A Receptor Is Required for β-Arrestin 2-mediated Activation of Extracellular Signal-regulated Kinases 1 and 2. J Biol Chem 2004; 279:48255-61. [PMID: 15355986 DOI: 10.1074/jbc.m406205200] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Binding of beta-arrestins to seven-membrane-spanning receptors (7MSRs) not only leads to receptor desensitization and endocytosis but also elicits additional signaling processes. We recently proposed that stimulation of the angiotensin type 1A (AT(1A)) receptor results in independent beta-arrestin 2- and G protein-mediated extracellular signal-regulated kinases 1 and 2 (ERK1/2) activation. Here we utilize two AT(1A) mutant receptors to study these independent pathways, one truncated at residue 324, thus removing all potential carboxyl-terminal phosphorylation sites, and the other bearing four mutations in the serine/threonine-rich clusters in the carboxyl terminus. As assessed by confocal microscopy, the two mutant receptors interacted with beta-arrestin 2-green fluorescent protein with much lower affinity than did the wild-type receptor. In addition, the mutant receptors more robustly stimulated G protein-mediated inositol phosphate production. Approximately one-half of the wild-type AT(1A) receptor-stimulated ERK1/2 activation was via a beta-arrestin 2-dependent pathway (suppressed by beta-arrestin 2 small interfering RNA), whereas the rest was mediated by a G protein-dependent pathway (suppressed by protein kinase C inhibitor). ERK1/2 activation by the mutant receptors was insensitive to beta-arrestin 2 small interfering RNA but was reduced more than 80% by a protein kinase C inhibitor. The biochemical consequences of ERK activation by the G protein and beta-arrestin 2-dependent pathways were also distinct. G-protein-mediated ERK activation enhanced the transcription of early growth response 1, whereas beta-arrestin 2-dependent ERK activation did not. In addition, stimulation of the truncated AT(1A) mutant receptor caused significantly greater early growth response 1 transcription than did the wild-type receptor. These findings demonstrate how the ability of receptors to interact with beta-arrestins determines both the mechanism of ERK activation as well as the physiological consequences of this activation.
Collapse
Affiliation(s)
- Huijun Wei
- Howard Hughes Medical Institute, Departments of Medicine and Biochemistry, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | |
Collapse
|
36
|
Heydorn A, Søndergaard BP, Ersbøll B, Holst B, Nielsen FC, Haft CR, Whistler J, Schwartz TW. A library of 7TM receptor C-terminal tails. Interactions with the proposed post-endocytic sorting proteins ERM-binding phosphoprotein 50 (EBP50), N-ethylmaleimide-sensitive factor (NSF), sorting nexin 1 (SNX1), and G protein-coupled receptor-associated sorting protein (GASP). J Biol Chem 2004; 279:54291-303. [PMID: 15452121 DOI: 10.1074/jbc.m406169200] [Citation(s) in RCA: 139] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Adaptor and scaffolding proteins determine the cellular targeting, the spatial, and thereby the functional association of G protein-coupled seven-transmembrane receptors with co-receptors, transducers, and downstream effectors and the adaptors determine post-signaling events such as receptor sequestration through interactions, mainly with the C-terminal intracellular tails of the receptors. A library of tails from 59 representative members of the super family of seven-transmembrane receptors was probed as glutathione S-transferase fusion proteins for interactions with four different adaptor proteins previously proposed to be involved in post-endocytotic sorting of receptors. Of the two proteins suggested to target receptors for recycling to the cell membrane, which is the route believed to be taken by a majority of receptors, ERM (ezrin-radixin-moesin)-binding phosphoprotein 50 (EBP50) bound only a single receptor tail, i.e. the beta(2)-adrenergic receptor, whereas N-ethylmaleimide-sensitive factor bound 11 of the tail-fusion proteins. Of the two proteins proposed to target receptors for lysosomal degradation, sorting nexin 1 (SNX1) bound 10 and the C-terminal domain of G protein-coupled receptor-associated sorting protein bound 23 of the 59 tail proteins. Surface plasmon resonance analysis of the binding kinetics of selected hits from the glutathione S-transferase pull-down experiments, i.e. the tails of the virally encoded receptor US28 and the delta-opioid receptor, confirmed the expected nanomolar affinities for interaction with SNX1. Truncations of the NK(1) receptor revealed that an extended binding epitope is responsible for the interaction with both SNX1 and G protein-coupled receptor-associated sorting protein as well as with N-ethylmaleimide-sensitive factor. It is concluded that the tail library provides useful information on the general importance of certain adaptor proteins, for example, in this case, ruling out EBP50 as being a broad spectrum-recycling adaptor.
Collapse
MESH Headings
- Amino Acid Sequence
- Carrier Proteins/metabolism
- Cell Membrane/chemistry
- Cell Membrane/metabolism
- Endocytosis
- Gene Deletion
- Glutathione Transferase/genetics
- Humans
- Lysosomes/metabolism
- Molecular Sequence Data
- Mutagenesis
- Peptide Fragments/chemistry
- Peptide Fragments/genetics
- Peptide Fragments/metabolism
- Peptide Library
- Phosphoproteins
- Receptors, Adrenergic, beta-2/chemistry
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/metabolism
- Receptors, Cell Surface/chemistry
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Opioid, delta/chemistry
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/metabolism
- Receptors, Tachykinin/chemistry
- Receptors, Tachykinin/genetics
- Receptors, Tachykinin/metabolism
- Recombinant Fusion Proteins/metabolism
- Sodium-Hydrogen Exchangers/metabolism
- Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins
- Surface Plasmon Resonance
- Vesicular Transport Proteins/metabolism
Collapse
Affiliation(s)
- Arne Heydorn
- Laboratory for Molecular Pharmacology, Department of Pharmacology, Panum Institute, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Roosterman D, Cottrell GS, Schmidlin F, Steinhoff M, Bunnett NW. Recycling and resensitization of the neurokinin 1 receptor. Influence of agonist concentration and Rab GTPases. J Biol Chem 2004; 279:30670-9. [PMID: 15128739 DOI: 10.1074/jbc.m402479200] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Substance P (SP) induces endocytosis and recycling of the neurokinin 1 receptor (NK1R) in endothelial cells and spinal neurons at sites of inflammation and pain, and it is thus important to understand the mechanism and function of receptor trafficking. We investigated how the SP concentration affects NK1R trafficking and determined the role of Rab GTPases in trafficking. NK1R trafficking was markedly influenced by the SP concentration. High SP (10 nM) induced translocation of the NK1R and beta-arrestin 1 to perinuclear sorting endosomes containing Rab5a, where NK1R remained for >60 min. Low SP (1 nM) induced translocation of the NK1R to early endosomes located immediately beneath the plasma membrane that also contained Rab5a and beta-arrestin 1, followed by rapid recycling of the NK1R. Overexpression of Rab5a promoted NK1R translocation to perinuclear sorting endosomes, whereas the GTP binding-deficient mutant Rab5aS34N caused retention of the NK1R in superficial early endosomes. NK1R translocated from superficial early endosomes to recycling endosomes containing Rab4a and Rab11a, and Rab11aS25N inhibited NK1R recycling. Rapid NK1R recycling coincided with resensitization of SP-induced Ca2+ mobilization and with the return of surface SP binding sites. Resensitization was minimally affected by inhibition of vacuolar H(+)-ATPase and phosphatases but was markedly suppressed by disruption of Rab4a and Rab11a. Thus, whereas beta-arrestins mediate NK1R endocytosis, Rab5a regulates translocation between early and sorting endosomes, and Rab4a and Rab11a regulate trafficking through recycling endosomes. We have thus identified a new function of Rab5a as a control protein for directing concentration-dependent trafficking of the NK1R into different intracellular compartments and obtained evidence that Rab4a and Rab11a contribute to G-protein-coupled receptor recycling from early endosomes.
Collapse
Affiliation(s)
- Dirk Roosterman
- Departments of Surgery and Physiology, University of California San Francisco, San Francisco, California 94143-0660, USA
| | | | | | | | | |
Collapse
|