1
|
Yamashita A, Ito Y, Osada M, Matsuda H, Hosono K, Tsujikawa K, Okamoto H, Amano H. RAMP1 Signaling Mitigates Acute Lung Injury by Distinctively Regulating Alveolar and Monocyte-Derived Macrophages. Int J Mol Sci 2024; 25:10107. [PMID: 39337592 PMCID: PMC11432488 DOI: 10.3390/ijms251810107] [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: 09/08/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening lung injury that induces cytokine hypersecretion. Receptor activity-modifying protein (RAMP) 1, a subunit of the calcitonin gene-related peptide (CGRP) receptor, regulates the production of cytokines. This study examined the role of RAMP1 signaling during lipopolysaccharide (LPS)-induced acute lung injury (ALI). LPS administration to wild-type (WT) mice depleted alveolar macrophages (AMs) and recruited monocyte-derived macrophages (MDMs) and neutrophils. RAMP1-deficient (RAMP1-/-) mice exhibited higher lung injury scores, cytokine levels, and cytokine-producing neutrophil infiltration. RAMP1-deficient AMs produced more cytokines in response to LPS than WT AMs. Adoptive transfer of RAMP1-deficient AMs to RAMP1-/- mice increased cytokine levels and neutrophil accumulation compared to the transfer of WT AMs. RAMP1-/- mice had reduced MDM recruitment and lower pro-inflammatory and reparative macrophage profiles. Cultured bone marrow (BM)-derived RAMP1-deficient macrophages stimulated with LPS showed decreased expression of pro-inflammatory and pro-repairing genes. CGRP administration to WT mice reduced cytokine production and neutrophil accumulation. These findings indicate that RAMP1 signaling mitigates LPS-induced ALI by inactivating AMs and promoting inflammatory and repair activities of MDMs. Targeting RAMP1 signaling presents a potential therapeutic approach for the treatment of ARDS.
Collapse
Affiliation(s)
- Atsushi Yamashita
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara 252-0373, Japan; (A.Y.); (Y.I.); (K.H.)
- Department of Anesthesiology, Kitasato University School of Medicine, Sagamihara 252-0374, Japan (H.O.)
| | - Yoshiya Ito
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara 252-0373, Japan; (A.Y.); (Y.I.); (K.H.)
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara 252-0374, Japan
| | - Mayuko Osada
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara 252-0373, Japan; (A.Y.); (Y.I.); (K.H.)
- Department of Emergency and Critical Care Medicine, Kitasato University School of Medicine, Sagamihara 252-0374, Japan
| | - Hiromi Matsuda
- Department of Anesthesiology, Kitasato University School of Medicine, Sagamihara 252-0374, Japan (H.O.)
| | - Kanako Hosono
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara 252-0373, Japan; (A.Y.); (Y.I.); (K.H.)
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara 252-0374, Japan
| | - Kazutake Tsujikawa
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan;
| | - Hirotsugu Okamoto
- Department of Anesthesiology, Kitasato University School of Medicine, Sagamihara 252-0374, Japan (H.O.)
| | - Hideki Amano
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara 252-0373, Japan; (A.Y.); (Y.I.); (K.H.)
- Department of Pharmacology, Kitasato University School of Medicine, Sagamihara 252-0374, Japan
| |
Collapse
|
2
|
Nelson-Maney NP, Bálint L, Beeson AL, Serafin DS, Kistner BM, Douglas ES, Siddiqui AH, Tauro AM, Caron KM. Meningeal lymphatic CGRP signaling governs pain via cerebrospinal fluid efflux and neuroinflammation in migraine models. J Clin Invest 2024; 134:e175616. [PMID: 38743922 PMCID: PMC11290972 DOI: 10.1172/jci175616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
Recently developed antimigraine therapeutics targeting calcitonin gene-related peptide (CGRP) signaling are effective, though their sites of activity remain elusive. Notably, the lymphatic vasculature is responsive to CGRP signaling, but whether meningeal lymphatic vessels (MLVs) contribute to migraine pathophysiology is unknown. Mice with lymphatic vasculature deficient in the CGRP receptor (CalcrliLEC mice) treated with nitroglycerin-mediated (NTG-mediated) chronic migraine exhibit reduced pain and light avoidance compared with NTG-treated littermate controls. Gene expression profiles of lymphatic endothelial cells (LECs) isolated from the meninges of Rpl22HA/+;Lyve1Cre RiboTag mice treated with NTG revealed increased MLV-immune interactions compared with cells from untreated mice. Interestingly, the relative abundance of mucosal vascular addressin cell adhesion molecule 1-interacting (MAdCAM1-interacting) CD4+ T cells was increased in the deep cervical lymph nodes of NTG-treated control mice but not in NTG-treated CalcrliLEC mice. Treatment of cultured hLECs with CGRP peptide in vitro induced vascular endothelial-cadherin (VE-cadherin) rearrangement and reduced functional permeability. Likewise, intra cisterna magna injection of CGRP caused rearrangement of VE-cadherin, decreased MLV uptake of cerebrospinal fluid (CSF), and impaired CSF drainage in control mice but not in CalcrliLEC mice. Collectively, these findings reveal a previously unrecognized role for lymphatics in chronic migraine, whereby CGRP signaling primes MLV-immune interactions and reduces CSF efflux.
Collapse
|
3
|
Ike E, Kawano T, Takahashi K, Miyasaka T, Takahashi T. Calcitonin Gene-Related peptide receptor antagonist suppresses allergic asthma responses via downregulation of group 2 innate lymphoid cells in mice. Int Immunopharmacol 2023; 122:110608. [PMID: 37441811 DOI: 10.1016/j.intimp.2023.110608] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/20/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023]
Abstract
Allergic asthma is caused by chronic inflammation and hyper-responsiveness of the airway and is thought to be mediated by adaptive T helper type 2 (Th2)-driven immunity. However, recent studies have demonstrated that neuropeptide calcitonin gene-related peptide (CGRP)-mediated activation of group 2 innate lymphoid cells (ILC2s) may contribute to the development of asthma pathogenesis. Here, we investigated the therapeutic effects of the systemic administration of rimegepant, a CGRP receptor antagonist, on allergic asthma. Hyperplasia of CGRP-immunoreactive pulmonary neuroendocrine cells (PNECs) was observed in ovalbumin (OVA)-induced asthmatic mice. Concomitant with this, we observed an increase in the content of total lung CGRP. Upon antigen challenge, the concentration of plasma CGRP was transiently upregulated, whereas CGRP immunoreactivity within PNECs was intensively downregulated, suggesting that PNECs were the most likely source of CGRP. When rimegepant was administered according to CGRP kinetics, it suppressed asthma phenotypes, including airway hyper-responsiveness, infiltration of inflammatory cells in bronchoalveolar lavage fluid (BALF), hyperplasia of mucus-producing cells, and production of the Th2 cytokine IL-5. Moreover, we observed a decrease in the number of ILC2s and their capacity for IL-5 release in the presence of IL-33 in rimegepant-treated mice. In the allergic asthma model, rimegepant suppressed the activation of ILC2s mediated by PNEC-derived CGRP and subsequently impaired adaptive Th2-driven immunity, which ameliorated asthmatic phenotypes. Thus, an anti-CGRP signal strategy to target ILC2 will be a novel and attractive approach for treating allergic asthma that is refractory to other treatments.
Collapse
Affiliation(s)
- Erina Ike
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558 Japan
| | - Tasuku Kawano
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558 Japan
| | - Kento Takahashi
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558 Japan
| | - Tomomitsu Miyasaka
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558 Japan
| | - Tomoko Takahashi
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558 Japan.
| |
Collapse
|
4
|
Martin-Almeida M, Perez-Garcia J, Herrera-Luis E, Rosa-Baez C, Gorenjak M, Neerincx AH, Sardón-Prado O, Toncheva AA, Harner S, Wolff C, Brandstetter S, Valletta E, Abdel-Aziz MI, Hashimoto S, Berce V, Corcuera-Elosegui P, Korta-Murua J, Buntrock-Döpke H, Vijverberg SJH, Verster JC, Kerssemakers N, Hedman AM, Almqvist C, Villar J, Kraneveld AD, Potočnik U, Kabesch M, der Zee AHMV, Pino-Yanes M, Consortium OBOTS. Epigenome-Wide Association Studies of the Fractional Exhaled Nitric Oxide and Bronchodilator Drug Response in Moderate-to-Severe Pediatric Asthma. Biomedicines 2023; 11:biomedicines11030676. [PMID: 36979655 PMCID: PMC10044864 DOI: 10.3390/biomedicines11030676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/14/2023] [Accepted: 02/18/2023] [Indexed: 02/25/2023] Open
Abstract
Asthma is the most prevalent pediatric chronic disease. Bronchodilator drug response (BDR) and fractional exhaled nitric oxide (FeNO) are clinical biomarkers of asthma. Although DNA methylation (DNAm) contributes to asthma pathogenesis, the influence of DNAm on BDR and FeNO is scarcely investigated. This study aims to identify DNAm markers in whole blood associated either with BDR or FeNO in pediatric asthma. We analyzed 121 samples from children with moderate-to-severe asthma. The association of genome-wide DNAm with BDR and FeNO has been assessed using regression models, adjusting for age, sex, ancestry, and tissue heterogeneity. Cross-tissue validation was assessed in 50 nasal samples. Differentially methylated regions (DMRs) and enrichment in traits and biological pathways were assessed. A false discovery rate (FDR) < 0.1 and a genome-wide significance threshold of p < 9 × 10−8 were used to control for false-positive results. The CpG cg12835256 (PLA2G12A) was genome-wide associated with FeNO in blood samples (coefficient= −0.015, p = 2.53 × 10−9) and nominally associated in nasal samples (coefficient = −0.015, p = 0.045). Additionally, three CpGs were suggestively associated with BDR (FDR < 0.1). We identified 12 and four DMRs associated with FeNO and BDR (FDR < 0.05), respectively. An enrichment in allergic and inflammatory processes, smoking, and aging was observed. We reported novel associations of DNAm markers associated with BDR and FeNO enriched in asthma-related processes.
Collapse
Affiliation(s)
- Mario Martin-Almeida
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), 38200 San Cristóbal de La Laguna, Spain
| | - Javier Perez-Garcia
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), 38200 San Cristóbal de La Laguna, Spain
| | - Esther Herrera-Luis
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), 38200 San Cristóbal de La Laguna, Spain
| | - Carlos Rosa-Baez
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), 38200 San Cristóbal de La Laguna, Spain
| | - Mario Gorenjak
- Center for Human Molecular Genetics and Pharmacogenomics, Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
| | - Anne H. Neerincx
- Department of Respiratory Medicine, Amsterdam University Medical Centres—Loc. AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Olaia Sardón-Prado
- Division of Pediatric Respiratory Medicine, Donostia University Hospital, 20014 San Sebastián, Spain
- Department of Pediatrics, University of the Basque Country (UPV/EHU), 48013 San Sebastián, Spain
| | - Antoaneta A. Toncheva
- Department of Pediatric Pneumology and Allergy, University Children’s Hospital Regensburg (KUNO) at the Hospital St. Hedwig of the Order of St. John, University of Regensburg, D-93049 Regensburg, Germany
| | - Susanne Harner
- Department of Pediatric Pneumology and Allergy, University Children’s Hospital Regensburg (KUNO) at the Hospital St. Hedwig of the Order of St. John, University of Regensburg, D-93049 Regensburg, Germany
| | - Christine Wolff
- Department of Pediatric Pneumology and Allergy, University Children’s Hospital Regensburg (KUNO) at the Hospital St. Hedwig of the Order of St. John, University of Regensburg, D-93049 Regensburg, Germany
| | - Susanne Brandstetter
- Department of Pediatric Pneumology and Allergy, University Children’s Hospital Regensburg (KUNO) at the Hospital St. Hedwig of the Order of St. John, University of Regensburg, D-93049 Regensburg, Germany
| | - Elisa Valletta
- Department of Pediatric Pneumology and Allergy, University Children’s Hospital Regensburg (KUNO) at the Hospital St. Hedwig of the Order of St. John, University of Regensburg, D-93049 Regensburg, Germany
| | - Mahmoud I. Abdel-Aziz
- Department of Respiratory Medicine, Amsterdam University Medical Centres—Loc. AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Clinical Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt
| | - Simone Hashimoto
- Department of Respiratory Medicine, Amsterdam University Medical Centres—Loc. AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Pediatric Respiratory Medicine, Emma Children’s Hospital, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands
| | - Vojko Berce
- Center for Human Molecular Genetics and Pharmacogenomics, Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
- Clinic of Pediatrics, University Medical Centre Maribor, 2000 Maribor, Slovenia
| | - Paula Corcuera-Elosegui
- Division of Pediatric Respiratory Medicine, Donostia University Hospital, 20014 San Sebastián, Spain
| | - Javier Korta-Murua
- Division of Pediatric Respiratory Medicine, Donostia University Hospital, 20014 San Sebastián, Spain
- Department of Pediatrics, University of the Basque Country (UPV/EHU), 48013 San Sebastián, Spain
| | - Heike Buntrock-Döpke
- Department of Pediatric Pneumology and Allergy, University Children’s Hospital Regensburg (KUNO) at the Hospital St. Hedwig of the Order of St. John, University of Regensburg, D-93049 Regensburg, Germany
| | - Susanne J. H. Vijverberg
- Department of Respiratory Medicine, Amsterdam University Medical Centres—Loc. AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Joris C. Verster
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands
- Centre for Human Psychopharmacology, Swinburne University, Melbourne, VIC 3122, Australia
| | - Nikki Kerssemakers
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Anna M Hedman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Solna, 171 77 Stockholm, Sweden
| | - Catarina Almqvist
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Solna, 171 77 Stockholm, Sweden
| | - Jesús Villar
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Multidisciplinary Organ Dysfunction Evaluation Research Network, Research Unit, Hospital Universitario Dr. Negrín, 35010 Las Palmas de Gran Canaria, Spain
| | - Aletta D. Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Uroš Potočnik
- Center for Human Molecular Genetics and Pharmacogenomics, Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
- Clinic of Pediatrics, University Medical Centre Maribor, 2000 Maribor, Slovenia
- Laboratory for Biochemistry, Molecular Biology, and Genomics, Faculty of Chemistry and Chemical Engineering, University of Maribor, 2000 Maribor, Slovenia
| | - Michael Kabesch
- Department of Pediatric Pneumology and Allergy, University Children’s Hospital Regensburg (KUNO) at the Hospital St. Hedwig of the Order of St. John, University of Regensburg, D-93049 Regensburg, Germany
| | - Anke H. Maitland-van der Zee
- Department of Respiratory Medicine, Amsterdam University Medical Centres—Loc. AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Pediatric Respiratory Medicine, Emma Children’s Hospital, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands
| | - Maria Pino-Yanes
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), 38200 San Cristóbal de La Laguna, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna (ULL), 38200 San Cristóbal de La Laguna, Spain
- Correspondence: ; Tel.: +34-9223-16502-6343
| | | |
Collapse
|
5
|
Cremin M, Schreiber S, Murray K, Tay EXY, Reardon C. The diversity of neuroimmune circuits controlling lung inflammation. Am J Physiol Lung Cell Mol Physiol 2023; 324:L53-L63. [PMID: 36410021 PMCID: PMC9829467 DOI: 10.1152/ajplung.00179.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 10/30/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
It is becoming increasingly appreciated that the nervous and immune systems communicate bidirectionally to regulate immunological outcomes in a variety of organs including the lung. Activation of neuronal signaling can be induced by inflammation, tissue damage, or pathogens to evoke or reduce immune cell activation in what has been termed a neuroimmune reflex. In the periphery, these reflexes include the cholinergic anti-inflammatory pathway, sympathetic reflex, and sensory nociceptor-immune cell pathways. Continual advances in neuroimmunology in peripheral organ systems have fueled small-scale clinical trials that have yielded encouraging results for a range of immunopathologies such as rheumatoid arthritis. Despite these successes, several limitations should give clinical investigators pause in the application of neural stimulation as a therapeutic for lung inflammation, especially if inflammation arises from a novel pathogen. In this review, the general mechanisms of each reflex, the evidence for these circuits in the control of lung inflammation, and the key knowledge gaps in our understanding of these neuroimmune circuits will be discussed. These limitations can be overcome not only through a better understanding of neuroanatomy but also through a systematic evaluation of stimulation parameters using immune activation in lung tissues as primary readouts. Our rapidly evolving understanding of the nervous and immune systems highlights the importance of communication between these cells in health and disease. This integrative approach has tremendous potential in the development of targeted therapeutics if specific challenges can be overcome.
Collapse
Affiliation(s)
- Michael Cremin
- Department of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California
| | - Sierra Schreiber
- Department of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California
| | - Kaitlin Murray
- Department of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California
| | - Emmy Xue Yun Tay
- Department of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California
| | - Colin Reardon
- Department of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, UC Davis, Davis, California
| |
Collapse
|
6
|
Kotliar IB, Lorenzen E, Schwenk JM, Hay DL, Sakmar TP. Elucidating the Interactome of G Protein-Coupled Receptors and Receptor Activity-Modifying Proteins. Pharmacol Rev 2023; 75:1-34. [PMID: 36757898 PMCID: PMC9832379 DOI: 10.1124/pharmrev.120.000180] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 09/27/2022] [Indexed: 12/13/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are known to interact with several other classes of integral membrane proteins that modulate their biology and pharmacology. However, the extent of these interactions and the mechanisms of their effects are not well understood. For example, one class of GPCR-interacting proteins, receptor activity-modifying proteins (RAMPs), comprise three related and ubiquitously expressed single-transmembrane span proteins. The RAMP family was discovered more than two decades ago, and since then GPCR-RAMP interactions and their functional consequences on receptor trafficking and ligand selectivity have been documented for several secretin (class B) GPCRs, most notably the calcitonin receptor-like receptor. Recent bioinformatics and multiplexed experimental studies suggest that GPCR-RAMP interactions might be much more widespread than previously anticipated. Recently, cryo-electron microscopy has provided high-resolution structures of GPCR-RAMP-ligand complexes, and drugs have been developed that target GPCR-RAMP complexes. In this review, we provide a summary of recent advances in techniques that allow the discovery of GPCR-RAMP interactions and their functional consequences and highlight prospects for future advances. We also provide an up-to-date list of reported GPCR-RAMP interactions based on a review of the current literature. SIGNIFICANCE STATEMENT: Receptor activity-modifying proteins (RAMPs) have emerged as modulators of many aspects of G protein-coupled receptor (GPCR)biology and pharmacology. The application of new methodologies to study membrane protein-protein interactions suggests that RAMPs interact with many more GPCRs than had been previously known. These findings, especially when combined with structural studies of membrane protein complexes, have significant implications for advancing GPCR-targeted drug discovery and the understanding of GPCR pharmacology, biology, and regulation.
Collapse
Affiliation(s)
- Ilana B Kotliar
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York (I.B.K., E.L., T.P.S.); Tri-Institutional PhD Program in Chemical Biology, New York, New York (I.B.K.); Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Solna, Sweden (J.M.S.); Department of Pharmacology and Toxicology, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin, New Zealand (D.L.H.); and Department of Neurobiology, Care Sciences and Society (NVS), Division for Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden (T.P.S.)
| | - Emily Lorenzen
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York (I.B.K., E.L., T.P.S.); Tri-Institutional PhD Program in Chemical Biology, New York, New York (I.B.K.); Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Solna, Sweden (J.M.S.); Department of Pharmacology and Toxicology, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin, New Zealand (D.L.H.); and Department of Neurobiology, Care Sciences and Society (NVS), Division for Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden (T.P.S.)
| | - Jochen M Schwenk
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York (I.B.K., E.L., T.P.S.); Tri-Institutional PhD Program in Chemical Biology, New York, New York (I.B.K.); Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Solna, Sweden (J.M.S.); Department of Pharmacology and Toxicology, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin, New Zealand (D.L.H.); and Department of Neurobiology, Care Sciences and Society (NVS), Division for Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden (T.P.S.)
| | - Debbie L Hay
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York (I.B.K., E.L., T.P.S.); Tri-Institutional PhD Program in Chemical Biology, New York, New York (I.B.K.); Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Solna, Sweden (J.M.S.); Department of Pharmacology and Toxicology, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin, New Zealand (D.L.H.); and Department of Neurobiology, Care Sciences and Society (NVS), Division for Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden (T.P.S.)
| | - Thomas P Sakmar
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York (I.B.K., E.L., T.P.S.); Tri-Institutional PhD Program in Chemical Biology, New York, New York (I.B.K.); Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Solna, Sweden (J.M.S.); Department of Pharmacology and Toxicology, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin, New Zealand (D.L.H.); and Department of Neurobiology, Care Sciences and Society (NVS), Division for Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden (T.P.S.)
| |
Collapse
|
7
|
Characterization of Antibodies against Receptor Activity-Modifying Protein 1 (RAMP1): A Cautionary Tale. Int J Mol Sci 2022; 23:ijms232416035. [PMID: 36555690 PMCID: PMC9787598 DOI: 10.3390/ijms232416035] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/02/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
Calcitonin gene-related peptide (CGRP) is a key component of migraine pathophysiology, yielding effective migraine therapeutics. CGRP receptors contain a core accessory protein subunit: receptor activity-modifying protein 1 (RAMP1). Understanding of RAMP1 expression is incomplete, partly due to the challenges in identifying specific and validated antibody tools. We profiled antibodies for immunodetection of RAMP1 using Western blotting, immunocytochemistry and immunohistochemistry, including using RAMP1 knockout mouse tissue. Most antibodies could detect RAMP1 in Western blotting and immunocytochemistry using transfected cells. Two antibodies (844, ab256575) could detect a RAMP1-like band in Western blots of rodent brain but not RAMP1 knockout mice. However, cross-reactivity with other proteins was evident for all antibodies. This cross-reactivity prevented clear conclusions about RAMP1 anatomical localization, as each antibody detected a distinct pattern of immunoreactivity in rodent brain. We cannot confidently attribute immunoreactivity produced by RAMP1 antibodies (including 844) to the presence of RAMP1 protein in immunohistochemical applications in brain tissue. RAMP1 expression in brain and other tissues therefore needs to be revisited using RAMP1 antibodies that have been comprehensively validated using multiple strategies to establish multiple lines of convincing evidence. As RAMP1 is important for other GPCR/ligand pairings, our results have broader significance beyond the CGRP field.
Collapse
|
8
|
Hwang DDJ, Lee SJ, Kim JH, Lee SM. The Role of Neuropeptides in Pathogenesis of Dry Dye. J Clin Med 2021; 10:4248. [PMID: 34575359 PMCID: PMC8471988 DOI: 10.3390/jcm10184248] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/07/2021] [Accepted: 09/14/2021] [Indexed: 12/29/2022] Open
Abstract
Neuropeptides are known as important mediators between the nervous and immune systems. Recently, the role of the corneal nerve in the pathogenesis of various ocular surface diseases, including dry eye disease, has been highlighted. Neuropeptides are thought to be important factors in the pathogenesis of dry eye disease, as suggested by the well-known role between the nervous and immune systems, and several recently published studies have elucidated the previously unknown pathogenic mechanisms involved in the role of the neuropeptides secreted from the corneal nerves in dry eye disease. Here, we reviewed the emerging concept of neurogenic inflammation as one of the pathogenic mechanisms of dry eye disease, the recent results of related studies, and the direction of future research.
Collapse
Affiliation(s)
- Daniel Duck-Jin Hwang
- Department of Ophthalmology, HanGil Eye Hospital, Incheon 21388, Korea;
- Department of Ophthalmology, College of Medicine, Catholic Kwandong University, Incheon 21388, Korea
| | - Seok-Jae Lee
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul 03080, Korea; (S.-J.L.); (J.-H.K.)
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Korea
| | - Jeong-Hun Kim
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul 03080, Korea; (S.-J.L.); (J.-H.K.)
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Korea
- Department of Ophthalmology, College of Medicine, Seoul National University, Seoul 03080, Korea
- Advanced Biomedical Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon 34141, Korea
| | - Sang-Mok Lee
- Department of Ophthalmology, HanGil Eye Hospital, Incheon 21388, Korea;
- Department of Ophthalmology, College of Medicine, Catholic Kwandong University, Incheon 21388, Korea
| |
Collapse
|
9
|
Xu C, Gulinello M, Frenette PS. Nociceptors protect sickle cell disease mice from vaso-occlusive episodes and chronic organ damage. J Exp Med 2021; 218:182184. [PMID: 33045060 PMCID: PMC7534906 DOI: 10.1084/jem.20200065] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 06/17/2020] [Accepted: 08/27/2020] [Indexed: 12/29/2022] Open
Abstract
Sickle cell disease (SCD) is a common hereditary hematologic disorder. SCD patients suffer from acute vaso-occlusive episodes (VOEs), chronic organ damage, and premature death, with few therapeutic options. Although severe pain is a major clinical manifestation of SCD, it remains unknown whether nociception plays a role in SCD pathogenesis. To address this question, we generated nociceptor-deficient SCD mice and found, unexpectedly, that the absence of nociception led to more severe and more lethal VOE, indicating that somatosensory nerves protect SCD mice from VOE. Mechanistically, the beneficial effects of sensory nerves were induced by the neuropeptide calcitonin gene–related peptide (CGRP), which acted on hematopoietic cells. Additionally, oral capsaicin consumption, which can activate somatosensory nerves by binding to TRPV1, dramatically alleviated acute VOE and significantly prevented chronic liver and kidney damage in SCD mice. Thus, the manipulation of nociception may provide a promising approach to treat SCD.
Collapse
Affiliation(s)
- Chunliang Xu
- The Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Maria Gulinello
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY
| | - Paul S Frenette
- The Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY.,Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
10
|
Pavón-Romero GF, Serrano-Pérez NH, García-Sánchez L, Ramírez-Jiménez F, Terán LM. Neuroimmune Pathophysiology in Asthma. Front Cell Dev Biol 2021; 9:663535. [PMID: 34055794 PMCID: PMC8155297 DOI: 10.3389/fcell.2021.663535] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/15/2021] [Indexed: 12/26/2022] Open
Abstract
Asthma is a chronic inflammation of lower airway disease, characterized by bronchial hyperresponsiveness. Type I hypersensitivity underlies all atopic diseases including allergic asthma. However, the role of neurotransmitters (NT) and neuropeptides (NP) in this disease has been less explored in comparison with inflammatory mechanisms. Indeed, the airway epithelium contains pulmonary neuroendocrine cells filled with neurotransmitters (serotonin and GABA) and neuropeptides (substance P[SP], neurokinin A [NKA], vasoactive intestinal peptide [VIP], Calcitonin-gene related peptide [CGRP], and orphanins-[N/OFQ]), which are released after allergen exposure. Likewise, the autonomic airway fibers produce acetylcholine (ACh) and the neuropeptide Y(NPY). These NT/NP differ in their effects; SP, NKA, and serotonin exert pro-inflammatory effects, whereas VIP, N/OFQ, and GABA show anti-inflammatory activity. However, CGPR and ACh have dual effects. For example, the ACh-M3 axis induces goblet cell metaplasia, extracellular matrix deposition, and bronchoconstriction; the CGRP-RAMP1 axis enhances Th2 and Th9 responses; and the SP-NK1R axis promotes the synthesis of chemokines in eosinophils, mast cells, and neutrophils. In contrast, the ACh-α7nAChR axis in ILC2 diminishes the synthesis of TNF-α, IL-1, and IL-6, attenuating lung inflammation whereas, VIP-VPAC1, N/OFQ-NOP axes cause bronchodilation and anti-inflammatory effects. Some NT/NP as 5-HT and NKA could be used as biomarkers to monitor asthma patients. In fact, the asthma treatment based on inhaled corticosteroids and anticholinergics blocks M3 and TRPV1 receptors. Moreover, the administration of experimental agents such as NK1R/NK2R antagonists and exogenous VIP decrease inflammatory mediators, suggesting that regulating the effects of NT/NP represents a potential novel approach for the treatment of asthma.
Collapse
Affiliation(s)
| | | | | | | | - Luis M. Terán
- Department of Immunogenetics and Allergy, Instituto Nacional Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| |
Collapse
|
11
|
Ochoa-Callejero L, García-Sanmartín J, Villoslada-Blanco P, Íñiguez M, Pérez-Matute P, Pujadas E, Fowkes ME, Brody R, Oteo JA, Martínez A. Circulating Levels of Calcitonin Gene-Related Peptide Are Lower in COVID-19 Patients. J Endocr Soc 2021; 5:bvaa199. [PMID: 33506161 PMCID: PMC7798995 DOI: 10.1210/jendso/bvaa199] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Indexed: 12/27/2022] Open
Abstract
Background To better understand the biology of COVID-19, we have explored the behavior of calcitonin gene-related peptide (CGRP), an angiogenic, vasodilating, and immune modulating peptide, in severe acute respiratory syndrome coronavirus 2 positive patients. Methods Levels of CGRP in the serum of 57 COVID-19 patients (24 asymptomatic, 23 hospitalized in the general ward, and 10 admitted to the intensive care unit) and healthy donors (n = 24) were measured by enzyme-linked immunosorbent assay (ELISA). In addition, to better understand the physiological consequences of the observed variations, we investigated by immunofluorescence the distribution of receptor activity modifying protein 1 (RAMP1), one of the components of the CGRP receptor, in autopsy lung specimens. Results CGRP levels were greatly decreased in COVID-19 patients (P < 0.001) when compared to controls, and there were no significant differences due to disease severity, sex, age, or comorbidities. We found that COVID-19 patients treated with proton pump inhibitors had lower levels of CGRP than other patients not taking this treatment (P = 0.001). RAMP1 immunoreactivity was found in smooth muscle cells of large blood vessels and the bronchial tree and in the airways´ epithelium. In COVID-19 samples, RAMP1 was also found in proliferating type II pneumocytes, a common finding in these patients. Conclusions The lower levels of CGRP should negatively impact the respiratory physiology of COVID-19 patients due to vasoconstriction, improper angiogenesis, less epithelial repair, and faulty immune response. Therefore, restoring CGRP levels in these patients may represent a novel therapeutic approach for COVID-19.
Collapse
Affiliation(s)
| | | | | | - María Íñiguez
- Infectious Diseases, Microbiota, and Metabolism Unit (CIBIR), Logroño, Spain
| | | | - Elisabet Pujadas
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mary E Fowkes
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rachel Brody
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - José A Oteo
- Infectious Diseases, Microbiota, and Metabolism Unit (CIBIR), Logroño, Spain.,Infectious Diseases Department, Hospital Universitario San Pedro, Logroño, Spain
| | - Alfredo Martínez
- Oncology Area, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| |
Collapse
|
12
|
Gao X, Zhang D, Xu C, Li H, Caron KM, Frenette PS. Nociceptive nerves regulate haematopoietic stem cell mobilization. Nature 2021; 589:591-596. [PMID: 33361809 PMCID: PMC7856173 DOI: 10.1038/s41586-020-03057-y] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 11/19/2020] [Indexed: 11/17/2022]
Abstract
Haematopoietic stem cells (HSCs) reside in specialized microenvironments in the bone marrow-often referred to as 'niches'-that represent complex regulatory milieux influenced by multiple cellular constituents, including nerves1,2. Although sympathetic nerves are known to regulate the HSC niche3-6, the contribution of nociceptive neurons in the bone marrow remains unclear. Here we show that nociceptive nerves are required for enforced HSC mobilization and that they collaborate with sympathetic nerves to maintain HSCs in the bone marrow. Nociceptor neurons drive granulocyte colony-stimulating factor (G-CSF)-induced HSC mobilization via the secretion of calcitonin gene-related peptide (CGRP). Unlike sympathetic nerves, which regulate HSCs indirectly via the niche3,4,6, CGRP acts directly on HSCs via receptor activity modifying protein 1 (RAMP1) and the calcitonin receptor-like receptor (CALCRL) to promote egress by activating the Gαs/adenylyl cyclase/cAMP pathway. The ingestion of food containing capsaicin-a natural component of chili peppers that can trigger the activation of nociceptive neurons-significantly enhanced HSC mobilization in mice. Targeting the nociceptive nervous system could therefore represent a strategy to improve the yield of HSCs for stem cell-based therapeutic agents.
Collapse
Affiliation(s)
- Xin Gao
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, New York, USA.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Dachuan Zhang
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, New York, USA.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA.,These authors contributed equally
| | - Chunliang Xu
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, New York, USA.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA.,These authors contributed equally
| | - Huihui Li
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, New York, USA.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Kathleen M. Caron
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Paul S. Frenette
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, New York, USA.,Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
13
|
Andreou AP, Fuccaro M, Lambru G. The role of erenumab in the treatment of migraine. Ther Adv Neurol Disord 2020; 13:1756286420927119. [PMID: 32523630 PMCID: PMC7257830 DOI: 10.1177/1756286420927119] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 04/23/2020] [Indexed: 01/03/2023] Open
Abstract
Calcitonin gene related peptide (CGRP) monoclonal antibodies (mAbs) have been the
first class of specifically developed preventive treatments for migraine.
Clinical trials data suggest superiority of the CGRP mAbs to placebo in terms of
prevention of migraine symptoms, migraine-specific quality of life and headache
related disability. Treatment-related side effects overall did not differ
significantly from placebo and discontinuation rate due to side effects has been
low across the clinical trials, perhaps in view of their peripheral mode of
action. Along with their route and frequency of administration, these novel
class of drugs may constitute an improvement compared with the established
arsenal of migraine treatments. Erenumab is a fully human antibody and the only
mAb acting on the CGRP pathway by blocking its receptor. It is the first of the
CGRP mAb class approved by the US Food and Drug Administration (May 2018) and
the European Medicines Agency (July 2018). Erenumab exists in two different
doses (70 mg and 140 mg) and it is administered with monthly subcutaneous
injections. This review summarises erenumab pharmacological characteristics,
clinical trials data, focusing on the potential role of this treatment in
clinical practice.
Collapse
Affiliation(s)
- Anna P Andreou
- The Headache Service, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Matteo Fuccaro
- Department of Neurology, Treviso Hospital, Treviso, Italy
| | - Giorgio Lambru
- The Headache Service, Guy's and St Thomas' NHS Foundation Trust, Westminster Bridge Road, London, SE1 7EH, UK
| |
Collapse
|
14
|
Serafin DS, Harris NR, Nielsen NR, Mackie DI, Caron KM. Dawn of a New RAMPage. Trends Pharmacol Sci 2020; 41:249-265. [PMID: 32115276 PMCID: PMC7236817 DOI: 10.1016/j.tips.2020.01.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/24/2020] [Accepted: 01/27/2020] [Indexed: 01/08/2023]
Abstract
Receptor activity-modifying proteins (RAMPs) interact with G-protein-coupled receptors (GPCRs) to modify their functions, imparting significant implications upon their physiological and therapeutic potentials. Resurging interest in identifying RAMP-GPCR interactions has recently been fueled by coevolution studies and orthogonal technological screening platforms. These new studies reveal previously unrecognized RAMP-interacting GPCRs, many of which expand beyond Class B GPCRs. The consequences of these interactions on GPCR function and physiology lays the foundation for new molecular therapeutic targets, as evidenced by the recent success of erenumab. Here, we highlight recent papers that uncovered novel RAMP-GPCR interactions, human RAMP-GPCR disease-causing mutations, and RAMP-related human pathologies, paving the way for a new era of RAMP-targeted drug development.
Collapse
Affiliation(s)
- D Stephen Serafin
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Natalie R Harris
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Natalie R Nielsen
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Duncan I Mackie
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
15
|
Tao B, Jiang L, Chen L. Aberrant expression of calcitonin gene-related peptide and its correlation with prognosis in severe childhood pneumonia. Clinics (Sao Paulo) 2020; 75:e1448. [PMID: 31994614 PMCID: PMC6970281 DOI: 10.6061/clinics/2020/e1448] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/16/2019] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES The purpose of this study was to evaluate the relationship between the serum levels of calcitonin gene-related peptide (CGRP) and the prognosis of pediatric patients with severe pneumonia. METHODS Children diagnosed with severe pneumonia (n=76) were stratified into the survival (n=58) and non-survival groups (n=18) according to their 28-day survival status and into the non-risk (n=51), risk (n=17) and high-risk (n=8) categories based on the pediatric critical illness score (PCIS). Demographic data and laboratory results were collected. Serum CGRP levels were determined by enzyme-linked immunosorbent assay (ELISA). A receiver operating characteristic (ROC) curve was generated to determine the cutoff score for high CGRP levels. RESULTS Serum CGRP levels were significantly higher in the survival group than in the non-survival group and were significantly higher in the non-risk group than in the risk and high-risk groups. The ROC curve for the prognostic potential of CGRP yielded a significant area under the curve (AUC) value with considerable sensitivity and specificity. CONCLUSION Our findings show that CGRP downregulation might be a diagnostic marker that predicts the prognosis and survival of children with severe pneumonia.
Collapse
Affiliation(s)
- Baoqin Tao
- Emergency Department, Children's Hospital affiliated to Zhengzhou University, Zhengzhou, 450000 China
| | - Lei Jiang
- Emergency Department, First Affiliated Hospital of Naval Medical University, Shanghai, 200433 China
| | - Liang Chen
- Department of Respiratory, Xiamen Chang Gung Hospital Xiamen, Fujian Province, 361028 China
- *Corresponding author. E-mail:
| |
Collapse
|
16
|
Xie J, Guo J, Kanwal Z, Wu M, Lv X, Ibrahim NA, Li P, Buabeid MA, Arafa ESA, Sun Q. Calcitonin and Bone Physiology: In Vitro, In Vivo, and Clinical Investigations. Int J Endocrinol 2020; 2020:3236828. [PMID: 32963524 PMCID: PMC7501564 DOI: 10.1155/2020/3236828] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/18/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022] Open
Abstract
Calcitonin was discovered as a peptide hormone that was known to reduce the calcium levels in the systemic circulation. This hypocalcemic effect is produced due to multiple reasons such as inhibition of bone resorption or suppression of calcium release from the bone. Thus, calcitonin was said as a primary regulator of the bone resorption process. This is the reason why calcitonin has been used widely in clinics for the treatment of bone disorders such as osteoporosis, hypercalcemia, and Paget's disease. However, presently calcitonin usage is declined due to the development of efficacious formulations of new drugs. Calcitonin gene-related peptides and several other peptides such as intermedin, amylin, and adrenomedullin (ADM) are categorized in calcitonin family. These peptides are known for the structural similarity with calcitonin. Aside from having a similar structure, these peptides have few overlapping biological activities and signal transduction action through related receptors. However, several other activities are also present that are peptide specific. In vitro and in vivo studies documented the posttreatment effects of calcitonin peptides, i.e., positive effect on bone osteoblasts and their formation and negative effect on osteoclasts and their resorption. The recent research studies carried out on genetically modified mice showed the inhibition of osteoclast activity by amylin, while astonishingly calcitonin plays its role by suppressing osteoblast and bone turnover. This article describes the review of the bone, the activity of the calcitonin family of peptides, and the link between them.
Collapse
Affiliation(s)
- Jingbo Xie
- Department of Orthopedics, Fengcheng People's Hospital, Fengcheng, Jiangxi 331100, China
| | - Jian Guo
- Department of the Second Orthopedics, Hongdu Hospital of Traditional Chinese Medicine Affiliated to Jiangxi University of Traditional Chinese Medicine, Nanchang Hongdu Traditional Chinese Medicine Hospital, Nanchang, Jiangxi 330008, China
| | | | - Mingzheng Wu
- Department of Orthopaedics, Pu'ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, China
| | - Xiangyang Lv
- Department of Orthopaedics, Xi'an International Medical Center Hospital, Xi'an, Shaanxi 710100, China
| | | | - Ping Li
- Department of Orthopaedics, Ya'an People's Hospital, Ya'an, Sichuan 625000, China
| | | | | | - Qingshan Sun
- Department of Orthopedics, The Third Hospital of Shandong Province, Jinan, Shandong 250031, China
| |
Collapse
|
17
|
Wallrapp A, Burkett PR, Riesenfeld SJ, Kim SJ, Christian E, Abdulnour REE, Thakore PI, Schnell A, Lambden C, Herbst RH, Khan P, Tsujikawa K, Xavier RJ, Chiu IM, Levy BD, Regev A, Kuchroo VK. Calcitonin Gene-Related Peptide Negatively Regulates Alarmin-Driven Type 2 Innate Lymphoid Cell Responses. Immunity 2019; 51:709-723.e6. [PMID: 31604686 PMCID: PMC7076585 DOI: 10.1016/j.immuni.2019.09.005] [Citation(s) in RCA: 157] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/19/2019] [Accepted: 09/09/2019] [Indexed: 12/22/2022]
Abstract
Neuroimmune interactions have emerged as critical modulators of allergic inflammation, and type 2 innate lymphoid cells (ILC2s) are an important cell type for mediating these interactions. Here, we show that ILC2s expressed both the neuropeptide calcitonin gene-related peptide (CGRP) and its receptor. CGRP potently inhibited alarmin-driven type 2 cytokine production and proliferation by lung ILC2s both in vitro and in vivo. CGRP induced marked changes in ILC2 expression programs in vivo and in vitro, attenuating alarmin-driven proliferative and effector responses. A distinct subset of ILCs scored highly for a CGRP-specific gene signature after in vivo alarmin stimulation, suggesting CGRP regulated this response. Finally, we observed increased ILC2 proliferation and type 2 cytokine production as well as exaggerated responses to alarmins in mice lacking the CGRP receptor. Together, these data indicate that endogenous CGRP is a critical negative regulator of ILC2 responses in vivo.
Collapse
Affiliation(s)
- Antonia Wallrapp
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Patrick R Burkett
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Samantha J Riesenfeld
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Se-Jin Kim
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Elena Christian
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Raja-Elie E Abdulnour
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Pratiksha I Thakore
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alexandra Schnell
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Conner Lambden
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Rebecca H Herbst
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Pavana Khan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Kazutake Tsujikawa
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Bruce D Levy
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Howard Hughes Medical Institute and Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
18
|
Nagashima H, Mahlakõiv T, Shih HY, Davis FP, Meylan F, Huang Y, Harrison OJ, Yao C, Mikami Y, Urban JF, Caron KM, Belkaid Y, Kanno Y, Artis D, O'Shea JJ. Neuropeptide CGRP Limits Group 2 Innate Lymphoid Cell Responses and Constrains Type 2 Inflammation. Immunity 2019; 51:682-695.e6. [PMID: 31353223 PMCID: PMC6801073 DOI: 10.1016/j.immuni.2019.06.009] [Citation(s) in RCA: 202] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/12/2019] [Accepted: 06/14/2019] [Indexed: 01/10/2023]
Abstract
Innate lymphocytes maintain tissue homeostasis at mucosal barriers, with group 2 innate lymphoid cells (ILC2s) producing type 2 cytokines and controlling helminth infection. While the molecular understanding of ILC2 responses has advanced, the complexity of microenvironmental factors impacting ILC2s is becoming increasingly apparent. Herein, we used single-cell analysis to explore the diversity of gene expression among lung lymphocytes during helminth infection. Following infection, we identified a subset of ILC2s that preferentially expressed Il5-encoding interleukin (IL)-5, together with Calca-encoding calcitonin gene-related peptide (CGRP) and its cognate receptor components. CGRP in concert with IL-33 and neuromedin U (NMU) supported IL-5 but constrained IL-13 expression and ILC2 proliferation. Without CGRP signaling, ILC2 responses and worm expulsion were enhanced. Collectively, these data point to CGRP as a context-dependent negative regulatory factor that shapes innate lymphocyte responses to alarmins and neuropeptides during type 2 innate immune responses.
Collapse
Affiliation(s)
- Hiroyuki Nagashima
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, NIH, Bethesda, MD 20892, USA
| | - Tanel Mahlakõiv
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Han-Yu Shih
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, NIH, Bethesda, MD 20892, USA
| | - Fred P Davis
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, NIH, Bethesda, MD 20892, USA
| | - Francoise Meylan
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, NIH, Bethesda, MD 20892, USA
| | - Yuefeng Huang
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Oliver J Harrison
- Metaorganism Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Chen Yao
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, NIH, Bethesda, MD 20892, USA
| | - Yohei Mikami
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, NIH, Bethesda, MD 20892, USA
| | - Joseph F Urban
- US Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics, and Immunology Laboratory, Beltsville, MD 20705-2350, USA
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Yuka Kanno
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, NIH, Bethesda, MD 20892, USA.
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA.
| | - John J O'Shea
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
19
|
Naot D, Musson DS, Cornish J. The Activity of Peptides of the Calcitonin Family in Bone. Physiol Rev 2019; 99:781-805. [PMID: 30540227 DOI: 10.1152/physrev.00066.2017] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Calcitonin was discovered over 50 yr ago as a new hormone that rapidly lowers circulating calcium levels. This effect is caused by the inhibition of calcium efflux from bone, as calcitonin is a potent inhibitor of bone resorption. Calcitonin has been in clinical use for conditions of accelerated bone turnover, including Paget's disease and osteoporosis; although in recent years, with the development of drugs that are more potent inhibitors of bone resorption, its use has declined. A number of peptides that are structurally similar to calcitonin form the calcitonin family, which currently includes calcitonin gene-related peptides (αCGRP and βCGRP), amylin, adrenomedullin, and intermedin. Apart from being structurally similar, the peptides signal through related receptors and have some overlapping biological activities, although other activities are peptide specific. In bone, in vitro studies and administration of the peptides to animals generally found inhibitory effects on osteoclasts and bone resorption and positive effects on osteoblasts and bone formation. Surprisingly, studies in genetically modified mice have demonstrated that the physiological role of calcitonin appears to be the inhibition of osteoblast activity and bone turnover, whereas amylin inhibits osteoclast activity. The review article focuses on the activities of peptides of the calcitonin family in bone and the challenges in understanding the relationship between the pharmacological effects and the physiological roles of these peptides.
Collapse
Affiliation(s)
- Dorit Naot
- Department of Medicine, University of Auckland , Auckland , New Zealand
| | - David S Musson
- Department of Medicine, University of Auckland , Auckland , New Zealand
| | - Jillian Cornish
- Department of Medicine, University of Auckland , Auckland , New Zealand
| |
Collapse
|
20
|
Davis RB, Ding S, Nielsen NR, Pawlak JB, Blakeney ES, Caron KM. Calcitonin-Receptor-Like Receptor Signaling Governs Intestinal Lymphatic Innervation and Lipid Uptake. ACS Pharmacol Transl Sci 2019; 2:114-121. [PMID: 32219216 DOI: 10.1021/acsptsci.8b00061] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Indexed: 02/06/2023]
Abstract
The absorption of dietary fat requires complex neuroendocrine-mediated regulation of chylomicron trafficking through enterocytes and intestinal lymphatic vessels. Calcitonin-receptor-like receptor (Calcrl) is a G protein-coupled receptor that can bind either a lymphangiogenic ligand adrenomedullin, with coreceptor RAMP2, or the neuropeptide CGRP, with coreceptor RAMP1. The extent to which this common GPCR controls lipid absorption via lymphatics or enteric innervation remains unclear. We used conditional and inducible genetic deletion of Calcrl in lymphatics to elucidate the pathophysiological consequences of this receptor pathway under conditions of high-fat diet. Inefficient absorption of dietary fat coupled with altered lymphatic endothelial junctions in Calcrl fl/fl /Prox1-CreER T2 mice results in excessive, transcellular lipid accumulation and abnormal enterocyte chylomicron processing and failure to gain weight. Interestingly, Calcrl fl/fl /Prox1-CreER T2 animals show reduced and disorganized mucosal and submucosal innervation. Consistently, mice with genetic loss of the CGRP coreceptor RAMP1 also displayed mucosal and submucosal innervation deficits, substantiating the CGRP-biased function of Calcrl in the neurolymphocrine axis. Thus, the common Calcrl receptor is a critical regulator of lipid absorption through its cell-specific functions in neurolymphocrine crosstalk.
Collapse
Affiliation(s)
- Reema B Davis
- Department of Cell Biology and Physiology, University of North Carolina Chapel Hill, 111 Mason Farm Road, 6312B Medical Biomolecular Research Building, CB#7545, Chapel Hill, North Carolina 27599-7545, United States
| | - Shengli Ding
- Department of Cell Biology and Physiology, University of North Carolina Chapel Hill, 111 Mason Farm Road, 6312B Medical Biomolecular Research Building, CB#7545, Chapel Hill, North Carolina 27599-7545, United States
| | - Natalie R Nielsen
- Department of Cell Biology and Physiology, University of North Carolina Chapel Hill, 111 Mason Farm Road, 6312B Medical Biomolecular Research Building, CB#7545, Chapel Hill, North Carolina 27599-7545, United States
| | - John B Pawlak
- Department of Cell Biology and Physiology, University of North Carolina Chapel Hill, 111 Mason Farm Road, 6312B Medical Biomolecular Research Building, CB#7545, Chapel Hill, North Carolina 27599-7545, United States
| | - Elizabeth S Blakeney
- Department of Cell Biology and Physiology, University of North Carolina Chapel Hill, 111 Mason Farm Road, 6312B Medical Biomolecular Research Building, CB#7545, Chapel Hill, North Carolina 27599-7545, United States
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina Chapel Hill, 111 Mason Farm Road, 6312B Medical Biomolecular Research Building, CB#7545, Chapel Hill, North Carolina 27599-7545, United States
| |
Collapse
|
21
|
Sui P, Wiesner DL, Xu J, Zhang Y, Lee J, Van Dyken S, Lashua A, Yu C, Klein BS, Locksley RM, Deutsch G, Sun X. Pulmonary neuroendocrine cells amplify allergic asthma responses. Science 2018; 360:eaan8546. [PMID: 29599193 PMCID: PMC6387886 DOI: 10.1126/science.aan8546] [Citation(s) in RCA: 270] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 02/11/2018] [Accepted: 03/21/2018] [Indexed: 12/14/2022]
Abstract
Pulmonary neuroendocrine cells (PNECs) are rare airway epithelial cells whose function is poorly understood. Here we show that Ascl1-mutant mice that have no PNECs exhibit severely blunted mucosal type 2 response in models of allergic asthma. PNECs reside in close proximity to group 2 innate lymphoid cells (ILC2s) near airway branch points. PNECs act through calcitonin gene-related peptide (CGRP) to stimulate ILC2s and elicit downstream immune responses. In addition, PNECs act through the neurotransmitter γ-aminobutyric acid (GABA) to induce goblet cell hyperplasia. The instillation of a mixture of CGRP and GABA in Ascl1-mutant airways restores both immune and goblet cell responses. In accordance, lungs from human asthmatics show increased PNECs. These findings demonstrate that the PNEC-ILC2 neuroimmunological modules function at airway branch points to amplify allergic asthma responses.
Collapse
Affiliation(s)
- Pengfei Sui
- Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Darin L Wiesner
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jinhao Xu
- Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Yan Zhang
- Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jinwoo Lee
- Department of Medicine, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Steven Van Dyken
- Department of Medicine, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Amber Lashua
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Chuyue Yu
- Zhiyuan College, Shanghai JiaoTong University, Shanghai, China
| | - Bruce S Klein
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Richard M Locksley
- Department of Medicine, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Gail Deutsch
- Department of Laboratories, Seattle Children's Hospital, University of Washington, Seattle, WA 98105, USA
| | - Xin Sun
- Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA.
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
22
|
Voisin T, Bouvier A, Chiu IM. Neuro-immune interactions in allergic diseases: novel targets for therapeutics. Int Immunol 2018; 29:247-261. [PMID: 28814067 DOI: 10.1093/intimm/dxx040] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 07/05/2017] [Indexed: 12/13/2022] Open
Abstract
Recent studies have highlighted an emerging role for neuro-immune interactions in mediating allergic diseases. Allergies are caused by an overactive immune response to a foreign antigen. The peripheral sensory and autonomic nervous system densely innervates mucosal barrier tissues including the skin, respiratory tract and gastrointestinal (GI) tract that are exposed to allergens. It is increasingly clear that neurons actively communicate with and regulate the function of mast cells, dendritic cells, eosinophils, Th2 cells and type 2 innate lymphoid cells in allergic inflammation. Several mechanisms of cross-talk between the two systems have been uncovered, with potential anatomical specificity. Immune cells release inflammatory mediators including histamine, cytokines or neurotrophins that directly activate sensory neurons to mediate itch in the skin, cough/sneezing and bronchoconstriction in the respiratory tract and motility in the GI tract. Upon activation, these peripheral neurons release neurotransmitters and neuropeptides that directly act on immune cells to modulate their function. Somatosensory and visceral afferent neurons release neuropeptides including calcitonin gene-related peptide, substance P and vasoactive intestinal peptide, which can act on type 2 immune cells to drive allergic inflammation. Autonomic neurons release neurotransmitters including acetylcholine and noradrenaline that signal to both innate and adaptive immune cells. Neuro-immune signaling may play a central role in the physiopathology of allergic diseases including atopic dermatitis, asthma and food allergies. Therefore, getting a better understanding of these cellular and molecular neuro-immune interactions could lead to novel therapeutic approaches to treat allergic diseases.
Collapse
Affiliation(s)
- Tiphaine Voisin
- Department of Microbiology and Immunobiology, Division of Immunology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Amélie Bouvier
- Department of Microbiology and Immunobiology, Division of Immunology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Isaac M Chiu
- Department of Microbiology and Immunobiology, Division of Immunology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| |
Collapse
|
23
|
Oetjen LK, Kim BS. Interactions of the immune and sensory nervous systems in atopy. FEBS J 2018; 285:3138-3151. [PMID: 29637705 DOI: 10.1111/febs.14465] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/20/2018] [Accepted: 04/03/2018] [Indexed: 12/15/2022]
Abstract
A striking feature underlying all atopic disorders, such as asthma, atopic dermatitis, and food allergy, is the presence of pathologic sensory responses, reflexes, and behaviors. These symptoms, exemplified by chronic airway irritation and cough, chronic itch and scratching, as well as gastrointestinal discomfort and dysfunction, are often cited as the most debilitating aspects of atopic disorders. Emerging studies have highlighted how the immune system shapes the scope and intensity of sensory responses by directly modulating the sensory nervous system. Additionally, factors produced by neurons have demonstrated novel functions in propagating atopic inflammation at barrier surfaces. In this review, we highlight new studies that have changed our understanding of atopy through advances in characterizing the reciprocal interactions between the immune and sensory nervous systems.
Collapse
Affiliation(s)
- Landon K Oetjen
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO, USA.,Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian S Kim
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO, USA.,Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.,Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
24
|
Wu T, Huang J, Moore PJ, Little MS, Walton WG, Fellner RC, Alexis NE, Peter Di Y, Redinbo MR, Tilley SL, Tarran R. Identification of BPIFA1/SPLUNC1 as an epithelium-derived smooth muscle relaxing factor. Nat Commun 2017; 8:14118. [PMID: 28165446 PMCID: PMC5303822 DOI: 10.1038/ncomms14118] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 11/30/2016] [Indexed: 01/02/2023] Open
Abstract
Asthma is a chronic airway disease characterized by inflammation, mucus hypersecretion and abnormal airway smooth muscle (ASM) contraction. Bacterial permeability family member A1, BPIFA1, is a secreted innate defence protein. Here we show that BPIFA1 levels are reduced in sputum samples from asthmatic patients and that BPIFA1 is secreted basolaterally from healthy, but not asthmatic human bronchial epithelial cultures (HBECs), where it suppresses ASM contractility by binding to and inhibiting the Ca2+ influx channel Orai1. We have localized this effect to a specific, C-terminal α-helical region of BPIFA1. Furthermore, tracheas from Bpifa1-/- mice are hypercontractile, and this phenotype is reversed by the addition of recombinant BPIFA1. Our data suggest that BPIFA1 deficiency in asthmatic airways promotes Orai1 hyperactivity, increased ASM contraction and airway hyperresponsiveness. Strategies that target Orai1 or the BPIFA1 deficiency in asthma may lead to novel therapies to treat this disease.
Collapse
Affiliation(s)
- Tongde Wu
- Cystic Fibrosis Center/Marsico Lung Institute, Marsico Hall, 125 Mason Farm Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA
| | - Julianne Huang
- Cystic Fibrosis Center/Marsico Lung Institute, Marsico Hall, 125 Mason Farm Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA.,Department of Chemistry, Genome Science Building, 250 Bell Tower Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA
| | - Patrick J Moore
- Cystic Fibrosis Center/Marsico Lung Institute, Marsico Hall, 125 Mason Farm Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA
| | - Michael S Little
- Department of Chemistry, Genome Science Building, 250 Bell Tower Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA
| | - William G Walton
- Department of Chemistry, Genome Science Building, 250 Bell Tower Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA
| | - Robert C Fellner
- Cystic Fibrosis Center/Marsico Lung Institute, Marsico Hall, 125 Mason Farm Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA
| | - Neil E Alexis
- Center for Environmental Medicine, Asthma, and Lung Biology, US EPA Human Studies Facility, 104 Mason Farm Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA
| | - Y Peter Di
- Department of Environmental and Occupational Health, University of Pittsburgh, 331 Bridgeside Point Building, Pittsburgh, Pennsylvania 15260, USA
| | - Matthew R Redinbo
- Department of Chemistry, Genome Science Building, 250 Bell Tower Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA
| | - Stephen L Tilley
- Cystic Fibrosis Center/Marsico Lung Institute, Marsico Hall, 125 Mason Farm Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA.,Center for Environmental Medicine, Asthma, and Lung Biology, US EPA Human Studies Facility, 104 Mason Farm Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA
| | - Robert Tarran
- Cystic Fibrosis Center/Marsico Lung Institute, Marsico Hall, 125 Mason Farm Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA.,Department of Cell Biology &Physiology, 5200 Medical Biomolecular Research Building, 111 Mason Farm Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA
| |
Collapse
|
25
|
Pawlak JB, Wetzel-Strong SE, Dunn MK, Caron KM. Cardiovascular effects of exogenous adrenomedullin and CGRP in Ramp and Calcrl deficient mice. Peptides 2017; 88:1-7. [PMID: 27940069 PMCID: PMC5706544 DOI: 10.1016/j.peptides.2016.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 11/28/2016] [Accepted: 12/07/2016] [Indexed: 11/16/2022]
Abstract
Adrenomedullin (AM) and calcitonin gene-related peptide (CGRP) are potent vasodilator peptides and serve as ligands for the G-protein coupled receptor (GPCR) calcitonin receptor-like receptor (CLR/Calcrl). Three GPCR accessory proteins called receptor activity-modifying proteins (RAMPs) modify the ligand binding affinity of the receptor such that the CLR/RAMP1 heterodimer preferably binds CGRP, while CLR/RAMP2 and CLR/RAMP3 have a stronger affinity for AM. Here we determine the contribution of each of the three RAMPs to blood pressure control in response to exogenous AM and CGRP by measuring the blood pressure of mice with genetic reduction or deletion of the receptor components. Thus, the cardiovascular response of Ramp1-/-, Ramp2+/-, Ramp3-/-, Ramp1-/-/Ramp3-/- double-knockout (dKO), and Calcrl+/- mice to AM and CGRP were compared to wildtype mice. While under anesthesia, Ramp1-/- male mice had significantly higher basal blood pressure than wildtype males; a difference which was not present in female mice. Additionally, anesthetized Ramp1-/-, Ramp3-/-, and Calcrl+/- male mice exhibited significantly higher basal blood pressure than females of the same genotype. The hypotensive response to intravenously injected AM was greatly attenuated in Ramp1-/- mice, and to a lesser extent in Ramp3-/- and Calcrl+/- mice. However, Ramp1-/-/Ramp3-/- dKO mice retained some hypotensive response to AM. These results suggest that the hypotensive effect of AM is primarily mediated through the CLR/RAMP1 heterodimer, but that AM signaling via CLR/RAMP2 and CLR/RAMP3 also contributes to some hypotensive action. On the other hand, CGRP's hypotensive activity seems to be predominantly through the CLR/RAMP1 heterodimer. With this knowledge, therapeutic AM or CGRP peptides could be designed to cause less hypotension while maintaining canonical receptor-RAMP mediated signaling.
Collapse
Affiliation(s)
- J B Pawlak
- Department of Cell Biology and Physiology, 111 Mason Farm Rd., 6312B MBRB CB# 7545, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - S E Wetzel-Strong
- Department of Cell Biology and Physiology, 111 Mason Farm Rd., 6312B MBRB CB# 7545, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - M K Dunn
- Ferring Research Institute, Inc., 4245 Sorrento Valley Blvd., San Diego, CA 92121, USA
| | - K M Caron
- Department of Cell Biology and Physiology, 111 Mason Farm Rd., 6312B MBRB CB# 7545, The University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
26
|
Wang F, Chen Z, Ren X, Tian Y, Wang F, Liu C, Jin P, Li Z, Zhang F, Zhu B. Hormone-sensitive lipase deficiency alters gene expression and cholesterol content of mouse testis. Reproduction 2016; 153:175-185. [PMID: 27920259 PMCID: PMC5148802 DOI: 10.1530/rep-16-0484] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 10/04/2016] [Accepted: 11/10/2016] [Indexed: 12/15/2022]
Abstract
Hormone-sensitive lipase-knockout (HSL−/−) mice exhibit azoospermia for unclear reasons. To explore the basis of sterility, we performed the following three experiments. First, HSL protein distribution in the testis was determined. Next, transcriptome analyses were performed on the testes of three experimental groups. Finally, the fatty acid and cholesterol levels in the testes with three different genotypes studied were determined. We found that the HSL protein was present from spermatocyte cells to mature sperm acrosomes in wild-type (HSL+/+) testes. Spermiogenesis ceased at the elongation phase of HSL−/− testes. Transcriptome analysis indicated that genes involved in lipid metabolism, cell membrane, reproduction and inflammation-related processes were disordered in HSL−/− testes. The cholesterol content was significantly higher in HSL−/− than that in HSL+/+ testis. Therefore, gene expression and cholesterol ester content differed in HSL−/− testes compared to other testes, which may explain the sterility of male HSL−/− mice.
Collapse
Affiliation(s)
- Feng Wang
- College of Life SciencesCapital Normal University, Beijing, China
| | - Zheng Chen
- College of Life SciencesCapital Normal University, Beijing, China
| | - Xiaofang Ren
- College of Life SciencesCapital Normal University, Beijing, China
| | - Ye Tian
- College of Life SciencesCapital Normal University, Beijing, China
| | - Fucheng Wang
- College of Life SciencesCapital Normal University, Beijing, China
| | - Chao Liu
- College of Life SciencesCapital Normal University, Beijing, China
| | - Pengcheng Jin
- College of Life SciencesCapital Normal University, Beijing, China
| | - Zongyue Li
- College of Life SciencesCapital Normal University, Beijing, China
| | - Feixiong Zhang
- College of Life SciencesCapital Normal University, Beijing, China
| | - Baochang Zhu
- College of Life SciencesCapital Normal University, Beijing, China
| |
Collapse
|
27
|
High M, Cho HY, Marzec J, Wiltshire T, Verhein KC, Caballero MT, Acosta PL, Ciencewicki J, McCaw ZR, Kobzik L, Miller-DeGraff L, Gladwell W, Peden DB, Serra ME, Shi M, Weinberg C, Suzuki O, Wang X, Bell DA, Polack FP, Kleeberger SR. Determinants of host susceptibility to murine respiratory syncytial virus (RSV) disease identify a role for the innate immunity scavenger receptor MARCO gene in human infants. EBioMedicine 2016; 11:73-84. [PMID: 27554839 PMCID: PMC5049919 DOI: 10.1016/j.ebiom.2016.08.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 08/02/2016] [Accepted: 08/05/2016] [Indexed: 11/29/2022] Open
Abstract
Background Respiratory syncytial virus (RSV) is the global leading cause of lower respiratory tract infection in infants. Nearly 30% of all infected infants develop severe disease including bronchiolitis, but susceptibility mechanisms remain unclear. Methods We infected a panel of 30 inbred strains of mice with RSV and measured changes in lung disease parameters 1 and 5 days post-infection and they were used in genome-wide association (GWA) studies to identify quantitative trait loci (QTL) and susceptibility gene candidates. Findings GWA identified QTLs for RSV disease phenotypes, and the innate immunity scavenger receptor Marco was a candidate susceptibility gene; targeted deletion of Marco worsened murine RSV disease. We characterized a human MARCO promoter SNP that caused loss of gene expression, increased in vitro cellular response to RSV infection, and associated with increased risk of disease severity in two independent populations of children infected with RSV. Interpretation Translational integration of a genetic animal model and in vitro human studies identified a role for MARCO in human RSV disease severity. Because no RSV vaccines are approved for clinical use, genetic studies have implications for diagnosing individuals who are at risk for severe RSV disease, and disease prevention strategies (e.g. RSV antibodies). In a panel of inbred strains of mice, RSV disease phenotypes were characterized that resemble those in human disease. We identified Marco as a susceptibility gene, and a human MARCO mutation increased risk of disease severity in children. These studies have implications for diagnosing individuals who are at risk for severe RSV disease and prevent disease.
RSV disease is the primary global cause for hospitalization one year after birth but the causes of differential RSV disease severity are not understood. We show that RSV disease phenotypes vary significantly between inbred strains of mice, and resemble those in human disease. We used genetic approaches to identify and validate the innate immunity gene Marco as a host susceptibility determinant for murine RSV disease. We then characterized a loss of function polymorphism in human MARCO that increases risk of severe RSV disease risk in infants. Results have important implications for identifying genetic risk factors for severe RSV disease.
Collapse
Affiliation(s)
- Monica High
- Immunity, Inflammation, and Diseases Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Hye-Youn Cho
- Immunity, Inflammation, and Diseases Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Jacqui Marzec
- Immunity, Inflammation, and Diseases Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Tim Wiltshire
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Kirsten C Verhein
- Immunity, Inflammation, and Diseases Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | | | - Patricio L Acosta
- Fundación INFANT, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Cientificas y Tecnicas (CONICET), Buenos Aires, Argentina
| | - Jonathan Ciencewicki
- Immunity, Inflammation, and Diseases Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Zackary R McCaw
- Immunity, Inflammation, and Diseases Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Lester Kobzik
- Department of Environmental Health, Harvard University School of Public Health, Boston, MA, USA
| | - Laura Miller-DeGraff
- Immunity, Inflammation, and Diseases Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Wes Gladwell
- Immunity, Inflammation, and Diseases Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - David B Peden
- Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Min Shi
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Clarice Weinberg
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Oscar Suzuki
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Xuting Wang
- Genome Integrity & Structural Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Douglas A Bell
- Genome Integrity & Structural Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Fernando P Polack
- Fundación INFANT, Buenos Aires, Argentina; Department of Pediatrics, Vanderbilt University, Nashville, TN, USA.
| | - Steven R Kleeberger
- Immunity, Inflammation, and Diseases Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA.
| |
Collapse
|
28
|
Kechele DO, Dunworth WP, Trincot CE, Wetzel-Strong SE, Li M, Ma H, Liu J, Caron KM. Endothelial Restoration of Receptor Activity-Modifying Protein 2 Is Sufficient to Rescue Lethality, but Survivors Develop Dilated Cardiomyopathy. Hypertension 2016; 68:667-77. [PMID: 27402918 DOI: 10.1161/hypertensionaha.116.07191] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 06/03/2016] [Indexed: 12/20/2022]
Abstract
RAMPs (receptor activity-modifying proteins) serve as oligomeric modulators for numerous G-protein-coupled receptors, yet elucidating the physiological relevance of these interactions remains complex. Ramp2 null mice are embryonic lethal, with cardiovascular developmental defects similar to those observed in mice null for canonical adrenomedullin/calcitonin receptor-like receptor signaling. We aimed to genetically rescue the Ramp2(-/-) lethality in order to further delineate the spatiotemporal requirements for RAMP2 function during development and thereby enable the elucidation of an expanded repertoire of RAMP2 functions with family B G-protein-coupled receptors in adult homeostasis. Endothelial-specific expression of Ramp2 under the VE-cadherin promoter resulted in the partial rescue of Ramp2(-/-) mice, demonstrating that endothelial expression of Ramp2 is necessary and sufficient for survival. The surviving Ramp2(-/-) Tg animals lived to adulthood and developed spontaneous hypotension and dilated cardiomyopathy, which was not observed in adult mice lacking calcitonin receptor-like receptor. Yet, the hearts of Ramp2(-/-) Tg animals displayed dysregulation of family B G-protein-coupled receptors, including parathyroid hormone and glucagon receptors, as well as their downstream signaling pathways. These data suggest a functional requirement for RAMP2 in the modulation of additional G-protein-coupled receptor pathways in vivo, which is critical for sustained cardiovascular homeostasis. The cardiovascular importance of RAMP2 extends beyond the endothelium and canonical adrenomedullin/calcitonin receptor-like receptor signaling, in which future studies could elucidate novel and pharmacologically tractable pathways for treating cardiovascular diseases.
Collapse
Affiliation(s)
- Daniel O Kechele
- From the Department of Cell Biology and Physiology (D.O.K., S.E.W.-S., M.L., K.M.C.), Curriculum in Genetics and Molecular Biology (W.P.D., C.E.T., K.M.C.), Department of Pathology and Laboratory Medicine (H.M., J.L.), and McAllister Heart Institute (H.M., J.L., K.M.C.), The University of North Carolina, Chapel Hill
| | - William P Dunworth
- From the Department of Cell Biology and Physiology (D.O.K., S.E.W.-S., M.L., K.M.C.), Curriculum in Genetics and Molecular Biology (W.P.D., C.E.T., K.M.C.), Department of Pathology and Laboratory Medicine (H.M., J.L.), and McAllister Heart Institute (H.M., J.L., K.M.C.), The University of North Carolina, Chapel Hill
| | - Claire E Trincot
- From the Department of Cell Biology and Physiology (D.O.K., S.E.W.-S., M.L., K.M.C.), Curriculum in Genetics and Molecular Biology (W.P.D., C.E.T., K.M.C.), Department of Pathology and Laboratory Medicine (H.M., J.L.), and McAllister Heart Institute (H.M., J.L., K.M.C.), The University of North Carolina, Chapel Hill
| | - Sarah E Wetzel-Strong
- From the Department of Cell Biology and Physiology (D.O.K., S.E.W.-S., M.L., K.M.C.), Curriculum in Genetics and Molecular Biology (W.P.D., C.E.T., K.M.C.), Department of Pathology and Laboratory Medicine (H.M., J.L.), and McAllister Heart Institute (H.M., J.L., K.M.C.), The University of North Carolina, Chapel Hill
| | - Manyu Li
- From the Department of Cell Biology and Physiology (D.O.K., S.E.W.-S., M.L., K.M.C.), Curriculum in Genetics and Molecular Biology (W.P.D., C.E.T., K.M.C.), Department of Pathology and Laboratory Medicine (H.M., J.L.), and McAllister Heart Institute (H.M., J.L., K.M.C.), The University of North Carolina, Chapel Hill
| | - Hong Ma
- From the Department of Cell Biology and Physiology (D.O.K., S.E.W.-S., M.L., K.M.C.), Curriculum in Genetics and Molecular Biology (W.P.D., C.E.T., K.M.C.), Department of Pathology and Laboratory Medicine (H.M., J.L.), and McAllister Heart Institute (H.M., J.L., K.M.C.), The University of North Carolina, Chapel Hill
| | - Jiandong Liu
- From the Department of Cell Biology and Physiology (D.O.K., S.E.W.-S., M.L., K.M.C.), Curriculum in Genetics and Molecular Biology (W.P.D., C.E.T., K.M.C.), Department of Pathology and Laboratory Medicine (H.M., J.L.), and McAllister Heart Institute (H.M., J.L., K.M.C.), The University of North Carolina, Chapel Hill
| | - Kathleen M Caron
- From the Department of Cell Biology and Physiology (D.O.K., S.E.W.-S., M.L., K.M.C.), Curriculum in Genetics and Molecular Biology (W.P.D., C.E.T., K.M.C.), Department of Pathology and Laboratory Medicine (H.M., J.L.), and McAllister Heart Institute (H.M., J.L., K.M.C.), The University of North Carolina, Chapel Hill.
| |
Collapse
|
29
|
Klein KR, Matson BC, Caron KM. The expanding repertoire of receptor activity modifying protein (RAMP) function. Crit Rev Biochem Mol Biol 2016; 51:65-71. [PMID: 26740457 DOI: 10.3109/10409238.2015.1128875] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Receptor activity modifying proteins (RAMPs) associate with G-protein-coupled receptors (GPCRs) at the plasma membrane and together bind a variety of peptide ligands, serving as a communication interface between the extracellular and intracellular environments. The collection of RAMP-interacting GPCRs continues to expand and now consists of GPCRs from families A, B and C, suggesting that RAMP activity is extremely prevalent. RAMP association with GPCRs can regulate GPCR function by altering ligand binding, receptor trafficking and desensitization, and downstream signaling pathways. Here, we elaborate on these RAMP-dependent mechanisms of GPCR regulation, which provide opportunities for pharmacological intervention.
Collapse
Affiliation(s)
| | | | - Kathleen M Caron
- a Department of Cell Biology & Physiology and.,b Department of Genetics , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| |
Collapse
|
30
|
Kashem SW, Riedl MS, Yao C, Honda CN, Vulchanova L, Kaplan DH. Nociceptive Sensory Fibers Drive Interleukin-23 Production from CD301b+ Dermal Dendritic Cells and Drive Protective Cutaneous Immunity. Immunity 2016; 43:515-26. [PMID: 26377898 DOI: 10.1016/j.immuni.2015.08.016] [Citation(s) in RCA: 316] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 06/30/2015] [Accepted: 07/28/2015] [Indexed: 12/24/2022]
Abstract
Innate resistance to Candida albicans in mucosal tissues requires the production of interleukin-17A (IL-17A) by tissue-resident cells early during infection, but the mechanism of cytokine production has not been precisely defined. In the skin, we found that dermal γδ T cells were the dominant source of IL-17A during C. albicans infection and were required for pathogen resistance. Induction of IL-17A from dermal γδ T cells and resistance to C. albicans required IL-23 production from CD301b(+) dermal dendritic cells (dDCs). In addition, we found that sensory neurons were directly activated by C. albicans. Ablation of sensory neurons increased susceptibility to C. albicans infection, which could be rescued by exogenous addition of the neuropeptide CGRP. These data define a model in which nociceptive pathways in the skin drive production of IL-23 by CD301b(+) dDCs resulting in IL-17A production from γδ T cells and resistance to cutaneous candidiasis.
Collapse
MESH Headings
- Animals
- Candida albicans/immunology
- Candida albicans/physiology
- Candidiasis/genetics
- Candidiasis/immunology
- Candidiasis/microbiology
- Cells, Cultured
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dermis/cytology
- Flow Cytometry
- Host-Pathogen Interactions/immunology
- Immunity/genetics
- Immunity/immunology
- Interleukin-17/genetics
- Interleukin-17/immunology
- Interleukin-17/metabolism
- Interleukin-23/genetics
- Interleukin-23/immunology
- Interleukin-23/metabolism
- Lectins, C-Type/immunology
- Lectins, C-Type/metabolism
- Mice, Inbred Strains
- Mice, Knockout
- Mice, Transgenic
- Oligonucleotide Array Sequence Analysis
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, Calcitonin Gene-Related Peptide/genetics
- Receptors, Calcitonin Gene-Related Peptide/immunology
- Receptors, Calcitonin Gene-Related Peptide/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sensory Receptor Cells/immunology
- Sensory Receptor Cells/metabolism
- Skin/immunology
- Skin/metabolism
- Skin/microbiology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Transcriptome/genetics
- Transcriptome/immunology
Collapse
Affiliation(s)
- Sakeen W Kashem
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Maureen S Riedl
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Chen Yao
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Christopher N Honda
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Lucy Vulchanova
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel H Kaplan
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
31
|
Martin VT, Fanning KM, Serrano D, Buse DC, Reed ML, Lipton RB. Asthma is a risk factor for new onset chronic migraine: Results from the American migraine prevalence and prevention study. Headache 2015; 56:118-31. [PMID: 26581563 DOI: 10.1111/head.12731] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2015] [Indexed: 01/01/2023]
Abstract
OBJECTIVES To test the hypothesis that in persons with episodic migraine (EM), asthma is a risk factor for the onset of chronic migraine (CM). BACKGROUND Migraine and asthma are comorbid chronic disorders with episodic attacks thought to involve inflammatory and neurological mechanisms. Herein, we assess the influence of asthma on the clinical course of EM. METHODS To be eligible for this observational cohort study, AMPP Study participants had to meet criteria for EM in 2008, complete the validated six-item asthma questionnaire from the European Community Respiratory Health Survey (ECRHS) in 2008, and provide follow-up data in 2009. Using the ECRHS, we defined asthma as a binary variable (present or absent) based on an empirical cut score and developed a Respiratory Symptom Severity Score (RSSS) based on the number of positive responses (no severity = 0 positive responses, low severity = 1-2 positive responses, moderate severity = 3-4 positive responses, high severity = 5-6 positive responses). Chronic migraine was the primary outcome measure and was defined as those with ≥15 headache days per month on the 2009 AMPP Study survey. We used logistic regression in separate models to assess the influence of asthma as a binary variable (Model 1) and RSSS score categories (Model 2 using no respiratory symptoms as the reference) on CM onset after adjusting for sociodemographic factors, headache day frequency, migraine preventive medication use, and medication overuse. RESULTS The eligible sample for this study included 4446 individuals with EM in 2008 of whom 17% had asthma. This group had a mean age of 50.4 and was 80.8% female. In 2009, new onset CM developed in 2.9% (131/4446) of the 2008 EM cohort, including 5.4% (40/746) of the asthma subgroup and 2.5% (91/3700) of the non-asthma subgroup. In comparison to those without asthma, the adjusted odds for individuals with asthma and EM in 2008 to develop CM in 2009 were greater than two (adjusted odds ratio [aOR] 2.1; 95% CI: 1.4-3.1). Using the RSSS, the aOR for CM onset increased with the number of asthma symptoms, but only those in the high RSSS category showed a statistically significant increase in the odds of chronic migraine onset in comparison with the no RSSS reference group (aOR 3.3; 95% CI 1.7-6.2). CONCLUSIONS Asthma is associated with an increased risk of new onset CM 1 year later among individuals with EM, with the highest risk being among those with the greatest number of respiratory symptoms. The exact mechanisms underlying this association are unknown, but could suggest mast cell degranulation, autonomic dysfunction, or shared genetic or environmental factors.
Collapse
Affiliation(s)
- Vincent T Martin
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | | | - Dawn C Buse
- Department of Neurology, Albert Einstein College of Medicine and Montefiore Headache Center, Bronx, NY, USA
| | | | - Richard B Lipton
- Department of Neurology, Albert Einstein College of Medicine and Montefiore Headache Center, Bronx, NY, USA.,Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
32
|
Abstract
It is now recognized that G protein-coupled receptors (GPCRs), once considered largely independent functional units, have a far more diverse molecular architecture. Receptor activity-modifying proteins (RAMPs) provide an important example of proteins that interact with GPCRs to modify their function. RAMPs are able to act as pharmacological switches and chaperones, and they can regulate signaling and/or trafficking in a receptor-dependent manner. This review covers recent discoveries in the RAMP field and summarizes the known GPCR partners and functions of RAMPs. We also discuss the first peptide-bound structures of RAMP-GPCR complexes, which give insight into the molecular mechanisms that enable RAMPs to alter the pharmacology and signaling of GPCRs.
Collapse
Affiliation(s)
- Debbie L Hay
- School of Biological Sciences and Maurice Wilkins Center, University of Auckland, Auckland 1142, New Zealand;
| | - Augen A Pioszak
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104;
| |
Collapse
|
33
|
van der Westhuizen ET, Valant C, Sexton PM, Christopoulos A. Endogenous Allosteric Modulators of G Protein–Coupled Receptors. J Pharmacol Exp Ther 2015; 353:246-60. [DOI: 10.1124/jpet.114.221606] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|