1
|
Zhao J, Hou Y, Yin C, Hu J, Gao T, Huang X, Zhang X, Xing J, An J, Wan S, Li J. Upregulation of histamine receptor H1 promotes tumor progression and contributes to poor prognosis in hepatocellular carcinoma. Oncogene 2019; 39:1724-1738. [PMID: 31740780 PMCID: PMC7033043 DOI: 10.1038/s41388-019-1093-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 10/25/2019] [Accepted: 10/29/2019] [Indexed: 12/27/2022]
Abstract
H1 histamine receptor (H1HR) belongs to the family of rhodopsin-like G-protein-coupled receptors. Recent studies have shown that H1HR expression is increased in several types of cancer. However, its functional roles in tumor progression remain largely unknown, especially in hepatocellular carcinoma (HCC). We found that H1HR is frequently unregulated in HCC, which is significantly associated with both recurrence-free survival and overall survival in HCC patients. Functional experiments revealed that H1HR promoted both the growth and metastasis of HCC cells by inducing cell cycle progression, formation of lamellipodia, production of matrix metalloproteinase 2, and suppression of cell apoptosis. Activation of cyclic adenosine monophosphate-dependent protein kinase A was found to be involved in H1HR-mediated HCC cell growth and metastasis. In addition, we found that overexpression of H1HR was mainly due to the downregulation of miR-940 in HCC cells. Moreover, the H1HR inhibitor terfenadine significantly suppressed tumor growth and metastasis in an HCC xenograft nude mice model. Our findings demonstrate that H1HR plays a critical role in the growth and metastasis of HCC cells, which provides experimental evidence supporting H1HR as a potential drug target for the treatment of HCC.
Collapse
Affiliation(s)
- Jing Zhao
- Center for Molecular Pathology, First Affiliated Hospital, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.,State Key Laboratory of Cancer Biology and Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Yiran Hou
- Medical College of Yan'an University, Yan'an, Shaanxi, 716000, China
| | - Chun Yin
- State Key Laboratory of Cancer Biology and Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Jing Hu
- State Key Laboratory of Cancer Biology and Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Tian Gao
- Department of Gynecology and Obstetrics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaojun Huang
- State Key Laboratory of Cancer Biology and Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaohong Zhang
- Department of Gynecology and Obstetrics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jinliang Xing
- State Key Laboratory of Cancer Biology and Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Jiaze An
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Shaogui Wan
- Center for Molecular Pathology, First Affiliated Hospital, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
| | - Jibin Li
- State Key Laboratory of Cancer Biology and Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
2
|
Razali NA, Nazarudin NA, Lai KS, Abas F, Ahmad S. Curcumin derivative, 2,6-bis(2-fluorobenzylidene)cyclohexanone (MS65) inhibits interleukin-6 production through suppression of NF-κB and MAPK pathways in histamine-induced human keratinocytes cell (HaCaT). BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 18:217. [PMID: 30012134 PMCID: PMC6048808 DOI: 10.1186/s12906-018-2223-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 04/27/2018] [Indexed: 12/29/2022]
Abstract
Background Histamine is a well-known mediator involved in skin allergic responses through up-regulation of pro-inflammatory cytokines. Antihistamines remain the mainstay of allergy treatment, but they were found limited in efficacy and associated with several common side effects. Therefore, alternative therapeutic preferences are derived from natural products in an effort to provide safe yet reliable anti-inflammatory agents. Curcumin and their derivatives are among compounds of interest in natural product research due to numerous pharmacological benefits including anti-inflammatory activities. Here, we investigate the effects of chemically synthesized curcumin derivative, 2,6-bis(2-fluorobenzylidene)cyclohexanone (MS65), in reducing cytokine production in histamine-induced HaCaT cells. Methods Interleukin (IL)-6 cytokine production in histamine-induced HaCaT cells were measured using enzyme-linked immunosorbent assay (ELISA) and cytotoxicity effects were determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Real-time polymerase chain reaction (RT-qPCR) was carried out to determine the inhibitory effects of MS65 on nuclear factor-kappa B (NF-κB) and mitogen activated protein kinase (MAPK) pathways. Results Histamine enhanced IL-6 production in HaCaT cells, with the highest production of IL-6 at 97.41 ± 2.33 pg/mL after 24 h of exposure. MS65 demonstrated a promising anti-inflammatory activity by inhibiting IL-6 production with half maximal inhibitory concentration (IC50) value of 4.91 ± 2.50 μM and median lethal concentration (LC50) value of 28.82 ± 7.56 μM. In gene expression level, we found that MS65 inhibits NF-κB and MAPK pathways through suppression of IKK/IκB/NFκB and c-Raf/MEK/ERK inflammatory cascades. Conclusion Taken together, our results suggest that MS65 could be used as a lead compound on developing new medicinal agent for the treatment of allergic skin diseases.
Collapse
|
3
|
Ganesh BP, Hall A, Ayyaswamy S, Nelson JW, Fultz R, Major A, Haag A, Esparza M, Lugo M, Venable S, Whary M, Fox JG, Versalovic J. Diacylglycerol kinase synthesized by commensal Lactobacillus reuteri diminishes protein kinase C phosphorylation and histamine-mediated signaling in the mammalian intestinal epithelium. Mucosal Immunol 2018; 11:380-393. [PMID: 28745328 PMCID: PMC5785580 DOI: 10.1038/mi.2017.58] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 05/21/2017] [Indexed: 02/04/2023]
Abstract
Lactobacillus reuteri 6475 (Lr) of the human microbiome synthesizes histamine and can suppress inflammation via type 2 histamine receptor (H2R) activation in the mammalian intestine. Gut microbes such as Lr promote H2R signaling and may suppress H1R proinflammatory signaling pathways in parallel by unknown mechanisms. In this study, we identified a soluble bacterial enzyme known as diacylglycerol kinase (Dgk) from Lr that is secreted into the extracellular milieu and presumably into the intestinal lumen. DgK diminishes diacylglycerol (DAG) quantities in mammalian cells by promoting its metabolic conversion and causing reduced protein kinase C phosphorylation (pPKC) as a net effect in mammalian cells. We demonstrated that histamine synthesized by gut microbes (Lr) activates both mammalian H1R and H2R, but Lr-derived Dgk suppresses the H1R signaling pathway. Phospho-PKC and IκBα were diminished within the intestinal epithelium of mice and humans treated by wild-type (WT) Lr, but pPKC and IκBα were not decreased in treatment with ΔdgkA Lr. Mucosal IL-6 and systemic interleukin (IL)-1α, eotaxin, and granulocyte colony-stimulating factor (G-CSF) were suppressed in WT Lr, but not in ΔdgkA Lr colonized mice. Collectively, the commensal microbe Lr may act as a "microbial antihistamine" by suppressing intestinal H1R-mediated proinflammatory responses via diminished pPKC-mediated mammalian cell signaling.
Collapse
Affiliation(s)
- Bhanu Priya Ganesh
- Departments of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
- Division of Comparative Medicine, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anne Hall
- Departments of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - Sriram Ayyaswamy
- Departments of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - James Willard Nelson
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Robert Fultz
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Angela Major
- Departments of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - Anthony Haag
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - Magdalena Esparza
- Departments of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - Monica Lugo
- Departments of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - Susan Venable
- Departments of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - Mark Whary
- Division of Comparative Medicine, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - James G. Fox
- Division of Comparative Medicine, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - James Versalovic
- Departments of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| |
Collapse
|
4
|
Tu L, Lu Z, Dieser K, Schmitt C, Chan SW, Ngan MP, Andrews PLR, Nalivaiko E, Rudd JA. Brain Activation by H 1 Antihistamines Challenges Conventional View of Their Mechanism of Action in Motion Sickness: A Behavioral, c-Fos and Physiological Study in Suncus murinus (House Musk Shrew). Front Physiol 2017; 8:412. [PMID: 28659825 PMCID: PMC5470052 DOI: 10.3389/fphys.2017.00412] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 05/30/2017] [Indexed: 12/21/2022] Open
Abstract
Motion sickness occurs under a variety of circumstances and is common in the general population. It is usually associated with changes in gastric motility, and hypothermia, which are argued to be surrogate markers for nausea; there are also reports that respiratory function is affected. As laboratory rodents are incapable of vomiting, Suncus murinus was used to model motion sickness and to investigate changes in gastric myoelectric activity (GMA) and temperature homeostasis using radiotelemetry, whilst also simultaneously investigating changes in respiratory function using whole body plethysmography. The anti-emetic potential of the highly selective histamine H1 receptor antagonists, mepyramine (brain penetrant), and cetirizine (non-brain penetrant), along with the muscarinic receptor antagonist, scopolamine, were investigated in the present study. On isolated ileal segments from Suncus murinus, both mepyramine and cetirizine non-competitively antagonized the contractile action of histamine with pK b values of 7.5 and 8.4, respectively; scopolamine competitively antagonized the contractile action of acetylcholine with pA2 of 9.5. In responding animals, motion (1 Hz, 4 cm horizontal displacement, 10 min) increased the percentage of the power of bradygastria, and decreased the percentage power of normogastria whilst also causing hypothermia. Animals also exhibited an increase in respiratory rate and a reduction in tidal volume. Mepyramine (50 mg/kg, i.p.) and scopolamine (10 mg/kg, i.p.), but not cetirizine (10 mg/kg, i.p.), significantly antagonized motion-induced emesis but did not reverse the motion-induced disruptions of GMA, or hypothermia, or effects on respiration. Burst analysis of plethysmographic-derived waveforms showed mepyramine also had increased the inter-retch+vomit frequency, and emetic episode duration. Immunohistochemistry demonstrated that motion alone did not induce c-fos expression in the brain. Paradoxically, mepyramine increased c-fos in brain areas regulating emesis control, and caused hypothermia; it also appeared to cause sedation and reduced the dominant frequency of slow waves. In conclusion, motion-induced emesis was associated with a disruption of GMA, respiration, and hypothermia. Mepyramine was a more efficacious anti-emetic than cetirizine, suggesting an important role of centrally-located H1 receptors. The ability of mepyramine to elevate c-fos provides a new perspective on how H1 receptors are involved in mechanisms of emesis control.
Collapse
Affiliation(s)
- Longlong Tu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China
| | - Zengbing Lu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China
| | - Karolina Dieser
- Department of Informatics and Microsystem Technology, University of Applied Sciences KaiserslauternZweibrücken, Germany
| | - Christina Schmitt
- Department of Informatics and Microsystem Technology, University of Applied Sciences KaiserslauternZweibrücken, Germany
| | - Sze Wa Chan
- School of Health Sciences, Caritas Institute of Higher EducationHong Kong, China
| | - Man P Ngan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China
| | - Paul L R Andrews
- Division of Biomedical Sciences, St. George's University of LondonLondon, United Kingdom
| | - Eugene Nalivaiko
- School of Biomedical Sciences and Pharmacy, University of NewcastleCallaghan, NSW, Australia
| | - John A Rudd
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong Kong, China.,Brain and Mind Institute, The Chinese University of Hong KongHong Kong, China
| |
Collapse
|
5
|
Haick JM, Brueggemann LI, Cribbs LL, Denning MF, Schwartz J, Byron KL. PKC-dependent regulation of Kv7.5 channels by the bronchoconstrictor histamine in human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2017; 312:L822-L834. [PMID: 28283479 DOI: 10.1152/ajplung.00567.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/23/2017] [Accepted: 03/07/2017] [Indexed: 12/13/2022] Open
Abstract
Kv7 potassium channels have recently been found to be expressed and functionally important for relaxation of airway smooth muscle. Previous research suggests that native Kv7 currents are inhibited following treatment of freshly isolated airway smooth muscle cells with bronchoconstrictor agonists, and in intact airways inhibition of Kv7 channels is sufficient to induce bronchiolar constriction. However, the mechanism by which Kv7 currents are inhibited by bronchoconstrictor agonists has yet to be elucidated. In the present study, native Kv7 currents in cultured human trachealis smooth muscle cells (HTSMCs) were observed to be inhibited upon treatment with histamine; inhibition of Kv7 currents was associated with membrane depolarization and an increase in cytosolic Ca2+ ([Ca2+]cyt). The latter response was inhibited by verapamil, a blocker of L-type voltage-sensitive Ca2+ channels (VSCCs). Protein kinase C (PKC) has been implicated as a mediator of bronchoconstrictor actions, although the targets of PKC are not clearly established. We found that histamine treatment significantly and dose-dependently suppressed currents through overexpressed wild-type human Kv7.5 (hKv7.5) channels in cultured HTSMCs, and this effect was inhibited by the PKC inhibitor Ro-31-8220 (3 µM). The PKC-dependent suppression of hKv7.5 currents corresponded with a PKC-dependent increase in hKv7.5 channel phosphorylation. Knocking down or inhibiting PKCα, or mutating hKv7.5 serine 441 to alanine, abolished the inhibitory effects of histamine on hKv7.5 currents. These findings provide the first evidence linking PKC activation to suppression of Kv7 currents, membrane depolarization, and Ca2+ influx via L-type VSCCs as a mechanism for histamine-induced bronchoconstriction.
Collapse
Affiliation(s)
- Jennifer M Haick
- Department of Molecular Pharmacology and Therapeutics, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Lioubov I Brueggemann
- Department of Molecular Pharmacology and Therapeutics, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Leanne L Cribbs
- Department of Cell & Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Mitchell F Denning
- Department of Pathology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois; and
| | - Jeffrey Schwartz
- Department of Thoracic and Cardiovascular Surgery, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Kenneth L Byron
- Department of Molecular Pharmacology and Therapeutics, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois;
| |
Collapse
|
6
|
Panula P, Chazot PL, Cowart M, Gutzmer R, Leurs R, Liu WLS, Stark H, Thurmond RL, Haas HL. International Union of Basic and Clinical Pharmacology. XCVIII. Histamine Receptors. Pharmacol Rev 2016; 67:601-55. [PMID: 26084539 DOI: 10.1124/pr.114.010249] [Citation(s) in RCA: 383] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Histamine is a developmentally highly conserved autacoid found in most vertebrate tissues. Its physiological functions are mediated by four 7-transmembrane G protein-coupled receptors (H1R, H2R, H3R, H4R) that are all targets of pharmacological intervention. The receptors display molecular heterogeneity and constitutive activity. H1R antagonists are long known antiallergic and sedating drugs, whereas the H2R was identified in the 1970s and led to the development of H2R-antagonists that revolutionized stomach ulcer treatment. The crystal structure of ligand-bound H1R has rendered it possible to design new ligands with novel properties. The H3R is an autoreceptor and heteroreceptor providing negative feedback on histaminergic and inhibition on other neurons. A block of these actions promotes waking. The H4R occurs on immuncompetent cells and the development of anti-inflammatory drugs is anticipated.
Collapse
Affiliation(s)
- Pertti Panula
- Department of Anatomy, and Neuroscience Center, University of Helsinki, Finland (P.P.); School of Biological and Biomedical Sciences, University of Durham, United Kingdom (P.L.C.); AbbVie, Inc. North Chicago, Illinois (M.C.); Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany (R.G.); Department of Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, The Netherlands (R.L.); Ziarco Pharma Limited, Canterbury, United Kingdom (W.L.S.L.); Institute of Pharmaceutical and Medical Chemistry and Institute of Neurophysiology, Medical Faculty, Westfalische-Wilhelms-University, Muenster, Germany (H.L.H.); Heinrich-Heine-University Duesseldorf, Germany (H.S.); and Janssen Research & Development, LLC, San Diego, California (R.L.T.)
| | - Paul L Chazot
- Department of Anatomy, and Neuroscience Center, University of Helsinki, Finland (P.P.); School of Biological and Biomedical Sciences, University of Durham, United Kingdom (P.L.C.); AbbVie, Inc. North Chicago, Illinois (M.C.); Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany (R.G.); Department of Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, The Netherlands (R.L.); Ziarco Pharma Limited, Canterbury, United Kingdom (W.L.S.L.); Institute of Pharmaceutical and Medical Chemistry and Institute of Neurophysiology, Medical Faculty, Westfalische-Wilhelms-University, Muenster, Germany (H.L.H.); Heinrich-Heine-University Duesseldorf, Germany (H.S.); and Janssen Research & Development, LLC, San Diego, California (R.L.T.)
| | - Marlon Cowart
- Department of Anatomy, and Neuroscience Center, University of Helsinki, Finland (P.P.); School of Biological and Biomedical Sciences, University of Durham, United Kingdom (P.L.C.); AbbVie, Inc. North Chicago, Illinois (M.C.); Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany (R.G.); Department of Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, The Netherlands (R.L.); Ziarco Pharma Limited, Canterbury, United Kingdom (W.L.S.L.); Institute of Pharmaceutical and Medical Chemistry and Institute of Neurophysiology, Medical Faculty, Westfalische-Wilhelms-University, Muenster, Germany (H.L.H.); Heinrich-Heine-University Duesseldorf, Germany (H.S.); and Janssen Research & Development, LLC, San Diego, California (R.L.T.)
| | - Ralf Gutzmer
- Department of Anatomy, and Neuroscience Center, University of Helsinki, Finland (P.P.); School of Biological and Biomedical Sciences, University of Durham, United Kingdom (P.L.C.); AbbVie, Inc. North Chicago, Illinois (M.C.); Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany (R.G.); Department of Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, The Netherlands (R.L.); Ziarco Pharma Limited, Canterbury, United Kingdom (W.L.S.L.); Institute of Pharmaceutical and Medical Chemistry and Institute of Neurophysiology, Medical Faculty, Westfalische-Wilhelms-University, Muenster, Germany (H.L.H.); Heinrich-Heine-University Duesseldorf, Germany (H.S.); and Janssen Research & Development, LLC, San Diego, California (R.L.T.)
| | - Rob Leurs
- Department of Anatomy, and Neuroscience Center, University of Helsinki, Finland (P.P.); School of Biological and Biomedical Sciences, University of Durham, United Kingdom (P.L.C.); AbbVie, Inc. North Chicago, Illinois (M.C.); Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany (R.G.); Department of Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, The Netherlands (R.L.); Ziarco Pharma Limited, Canterbury, United Kingdom (W.L.S.L.); Institute of Pharmaceutical and Medical Chemistry and Institute of Neurophysiology, Medical Faculty, Westfalische-Wilhelms-University, Muenster, Germany (H.L.H.); Heinrich-Heine-University Duesseldorf, Germany (H.S.); and Janssen Research & Development, LLC, San Diego, California (R.L.T.)
| | - Wai L S Liu
- Department of Anatomy, and Neuroscience Center, University of Helsinki, Finland (P.P.); School of Biological and Biomedical Sciences, University of Durham, United Kingdom (P.L.C.); AbbVie, Inc. North Chicago, Illinois (M.C.); Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany (R.G.); Department of Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, The Netherlands (R.L.); Ziarco Pharma Limited, Canterbury, United Kingdom (W.L.S.L.); Institute of Pharmaceutical and Medical Chemistry and Institute of Neurophysiology, Medical Faculty, Westfalische-Wilhelms-University, Muenster, Germany (H.L.H.); Heinrich-Heine-University Duesseldorf, Germany (H.S.); and Janssen Research & Development, LLC, San Diego, California (R.L.T.)
| | - Holger Stark
- Department of Anatomy, and Neuroscience Center, University of Helsinki, Finland (P.P.); School of Biological and Biomedical Sciences, University of Durham, United Kingdom (P.L.C.); AbbVie, Inc. North Chicago, Illinois (M.C.); Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany (R.G.); Department of Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, The Netherlands (R.L.); Ziarco Pharma Limited, Canterbury, United Kingdom (W.L.S.L.); Institute of Pharmaceutical and Medical Chemistry and Institute of Neurophysiology, Medical Faculty, Westfalische-Wilhelms-University, Muenster, Germany (H.L.H.); Heinrich-Heine-University Duesseldorf, Germany (H.S.); and Janssen Research & Development, LLC, San Diego, California (R.L.T.)
| | - Robin L Thurmond
- Department of Anatomy, and Neuroscience Center, University of Helsinki, Finland (P.P.); School of Biological and Biomedical Sciences, University of Durham, United Kingdom (P.L.C.); AbbVie, Inc. North Chicago, Illinois (M.C.); Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany (R.G.); Department of Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, The Netherlands (R.L.); Ziarco Pharma Limited, Canterbury, United Kingdom (W.L.S.L.); Institute of Pharmaceutical and Medical Chemistry and Institute of Neurophysiology, Medical Faculty, Westfalische-Wilhelms-University, Muenster, Germany (H.L.H.); Heinrich-Heine-University Duesseldorf, Germany (H.S.); and Janssen Research & Development, LLC, San Diego, California (R.L.T.)
| | - Helmut L Haas
- Department of Anatomy, and Neuroscience Center, University of Helsinki, Finland (P.P.); School of Biological and Biomedical Sciences, University of Durham, United Kingdom (P.L.C.); AbbVie, Inc. North Chicago, Illinois (M.C.); Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany (R.G.); Department of Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, The Netherlands (R.L.); Ziarco Pharma Limited, Canterbury, United Kingdom (W.L.S.L.); Institute of Pharmaceutical and Medical Chemistry and Institute of Neurophysiology, Medical Faculty, Westfalische-Wilhelms-University, Muenster, Germany (H.L.H.); Heinrich-Heine-University Duesseldorf, Germany (H.S.); and Janssen Research & Development, LLC, San Diego, California (R.L.T.)
| |
Collapse
|
7
|
Baker JG, Proudman RGW, Hill SJ. Impact of polymorphic variants on the molecular pharmacology of the two-agonist conformations of the human β1-adrenoceptor. PLoS One 2013; 8:e77582. [PMID: 24250787 PMCID: PMC3826719 DOI: 10.1371/journal.pone.0077582] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 09/04/2013] [Indexed: 11/18/2022] Open
Abstract
β-blockers are widely used to improve symptoms and prolong life in heart disease primarily by inhibiting the actions of endogenous catecholamines at the β1-adrenoceptor. There are two common naturally occurring polymorphisms within the human β1-adrenoceptor sequence: Ser or Gly at position 49 in the N-terminus and Gly or Arg at position 389 in the C-terminus and some clinical studies have suggested that expression of certain variants may be associated with disease and affect response to treatment with β-blockers. The β1-adrenoceptor also exists in two agonist conformations - a high affinity catecholamine conformation and a low affinity secondary agonist conformation. Receptor-effector coupling and intracellular signalling from the different conformations may be affected by the polymorphic variants. Here, we examine in detail the molecular pharmacology of the β1-adrenoceptor polymorphic variants with respect to ligand affinity, efficacy, activation of the different agonist conformations and signal transduction and determine whether the polymorphic variants do indeed affect this secondary conformation. Stable cell lines expressing the wildtype and polymorphic variants were constructed and receptor pharmacology examined using whole cell binding and intracellular secondary messenger techniques. There was no difference in affinity for agonists and antagonists at the human wildtype β1-adrenoceptor (Ser49/Gly389) and the polymorphic variants Gly49/Gly389 and Ser49/Arg389. Furthermore, the polymorphic variant receptors both have two active agonist conformations with pharmacological properties similar to the wildtype receptor. Although the polymorphism at position 389 is thought to occur in an intracellular domain important for Gs-coupling, the two agonist conformations of the polymorphic variants stimulate intracellular signalling pathways, including Gs-cAMP intracellular signalling, in a manner very similar to that of the wildtype receptor.
Collapse
Affiliation(s)
- Jillian G. Baker
- Cell Signalling, School of Life Sciences, University of Nottingham, Nottingham, Nottinghamshire, United Kingdom
| | - Richard G. W. Proudman
- Cell Signalling, School of Life Sciences, University of Nottingham, Nottingham, Nottinghamshire, United Kingdom
| | - Stephen J. Hill
- Cell Signalling, School of Life Sciences, University of Nottingham, Nottingham, Nottinghamshire, United Kingdom
| |
Collapse
|
8
|
Mizuguchi H, Terao T, Kitai M, Ikeda M, Yoshimura Y, Das AK, Kitamura Y, Takeda N, Fukui H. Involvement of protein kinase Cdelta/extracellular signal-regulated kinase/poly(ADP-ribose) polymerase-1 (PARP-1) signaling pathway in histamine-induced up-regulation of histamine H1 receptor gene expression in HeLa cells. J Biol Chem 2011; 286:30542-30551. [PMID: 21730054 DOI: 10.1074/jbc.m111.253104] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The histamine H(1) receptor (H1R) gene is up-regulated in patients with allergic rhinitis. However, the mechanism and reason underlying this up-regulation are still unknown. Recently, we reported that the H1R expression level is strongly correlated with the severity of allergic symptoms. Therefore, understanding the mechanism of this up-regulation will help to develop new anti-allergic drugs targeted for H1R gene expression. Here we studied the molecular mechanism of H1R up-regulation in HeLa cells that express H1R endogenously in response to histamine and phorbol 12-myristate 13-acetate (PMA). In HeLa cells, histamine stimulation caused up-regulation of H1R gene expression. Rottlerin, a PKCδ-selective inhibitor, inhibited up-regulation of H1R gene expression, but Go6976, an inhibitor of Ca(2+)-dependent PKCs, did not. Histamine or PMA stimulation resulted in PKCδ phosphorylation at Tyr(311) and Thr(505). Activation of PKCδ by H(2)O(2) resulted in H1R mRNA up-regulation. Overexpression of PKCδ enhanced up-regulation of H1R gene expression, and knockdown of the PKCδ gene suppressed this up-regulation. Histamine or PMA caused translocation PKCδ from the cytosol to the Golgi. U0126, an MEK inhibitor, and DPQ, a poly(ADP-ribose) polymerase-1 inhibitor, suppressed PMA-induced up-regulation of H1R gene expression. These results were confirmed by a luciferase assay using the H1R promoter. Phosphorylation of ERK and Raf-1 in response to PMA was also observed. However, real-time PCR analysis showed no inhibition of H1R mRNA up-regulation by a Raf-1 inhibitor. These results suggest the involvement of the PKCδ/ERK/poly(ADP-ribose) polymerase-1 signaling pathway in histamine- or PMA-induced up-regulation of H1R gene expression in HeLa cells.
Collapse
Affiliation(s)
| | - Takuma Terao
- Departments of Molecular Pharmacology, Tokushima 770-8505, Japan
| | - Mika Kitai
- Departments of Molecular Pharmacology, Tokushima 770-8505, Japan
| | - Mitsuhiro Ikeda
- Departments of Molecular Pharmacology, Tokushima 770-8505, Japan
| | | | - Asish Kumar Das
- Departments of Molecular Pharmacology, Tokushima 770-8505, Japan
| | - Yoshiaki Kitamura
- Otolaryngology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima 770-8505, Japan
| | - Noriaki Takeda
- Otolaryngology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima 770-8505, Japan
| | - Hiroyuki Fukui
- Departments of Molecular Pharmacology, Tokushima 770-8505, Japan.
| |
Collapse
|
9
|
Jurič DM, Mele T, Čarman-Kržan M. Involvement of histaminergic receptor mechanisms in the stimulation of NT-3 synthesis in astrocytes. Neuropharmacology 2011; 60:1309-17. [DOI: 10.1016/j.neuropharm.2011.01.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2010] [Revised: 01/10/2011] [Accepted: 01/11/2011] [Indexed: 12/12/2022]
|
10
|
Notcovich C, Diez F, Tubio MR, Baldi A, Kazanietz MG, Davio C, Shayo C. Histamine acting on H1 receptor promotes inhibition of proliferation via PLC, RAC, and JNK-dependent pathways. Exp Cell Res 2010; 316:401-11. [DOI: 10.1016/j.yexcr.2009.11.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Revised: 11/04/2009] [Accepted: 11/05/2009] [Indexed: 01/06/2023]
|
11
|
Rico S, Antonijoan RM, Barbanoj MJ. Ebastine in the light of CONGA recommendations for the development of third-generation antihistamines. J Asthma Allergy 2009; 2:73-92. [PMID: 21437146 PMCID: PMC3048600 DOI: 10.2147/jaa.s3108] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2009] [Indexed: 11/27/2022] Open
Abstract
In 2003 a consensus group on new-generation antihistamines (CONGA) defined the characteristics required for a third-generation H(1) antihistamine as there had been much controversy about this issue since the early 1990s. One of the antihistamines that had been claimed to belong to such a group is the second-generation antihistamine, ebastine. The objective of this review is to analyze the pharmacology of ebastine, in light of the CONGA recommendations for the development of new-generation antihistamines: (1) anti-inflammatory properties, (2) potency, efficacy and effectiveness, (3) lack of cardiotoxicity, (4) lack of drug interactions, (5) lack of CNS effects, and (6) pharmacological approach. Ebastine seems to have anti-inflammatory properties that help to ameliorate nasal congestion, though this has not yet been conclusively demonstrated. Its pharmacological-therapeutic profile does not differ greatly from that of other second-generation antihistamines. Its cardiac safety has been widely assessed and no cardiac toxicity has been found at therapeutic doses despite initial concerns. The risk of potentially relevant drug interactions has been investigated and ruled out. Ebastine does not produce sedation at therapeutic doses and drug interaction studies with classical CNS depressants have not demonstrated a synergistic effect. Pharmacologically, ebastine is an H(1) inverse agonist. Perhaps the answer to the quest for new-generation antihistamines lies not only in H(1) but in a combined approach with other histamine receptors.
Collapse
Affiliation(s)
- S Rico
- Centre d’Investigació de Medicaments, Institut de Recerca; Servei de Farmacologia Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Departament de Farmacologia i Terapèutica, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - RM Antonijoan
- Centre d’Investigació de Medicaments, Institut de Recerca; Servei de Farmacologia Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, Spain
| | - MJ Barbanoj
- Centre d’Investigació de Medicaments, Institut de Recerca; Servei de Farmacologia Clínica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Departament de Farmacologia i Terapèutica, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, Spain
| |
Collapse
|
12
|
Roumestan C, Henriquet C, Gougat C, Michel A, Bichon F, Portet K, Jaffuel D, Mathieu M. Histamine H1-receptor antagonists inhibit nuclear factor-kappaB and activator protein-1 activities via H1-receptor-dependent and -independent mechanisms. Clin Exp Allergy 2008; 38:947-56. [DOI: 10.1111/j.1365-2222.2008.02990.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
13
|
Noubade R, Milligan G, Zachary JF, Blankenhorn EP, del Rio R, Rincon M, Teuscher C. Histamine receptor H1 is required for TCR-mediated p38 MAPK activation and optimal IFN-gamma production in mice. J Clin Invest 2008; 117:3507-18. [PMID: 17965772 DOI: 10.1172/jci32792] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2007] [Accepted: 08/29/2007] [Indexed: 01/14/2023] Open
Abstract
Histamine receptor H1 (H1R) is a susceptibility gene in both experimental autoimmune encephalomyelitis (EAE) and experimental autoimmune orchitis (EAO), 2 classical T cell-mediated models of organ-specific autoimmune disease. Here we showed that expression of H1R in naive CD4+ T cells was required for maximal IFN-gamma production but was dispensable for proliferation. Moreover, H1R signaling at the time of TCR ligation was required for activation of p38 MAPK, a known regulator of IFN-gamma expression. Importantly, selective reexpression of H1R in CD4+ T cells fully complemented both the IFN-gamma production and the EAE susceptibility of H1R-deficient mice. These data suggest that the presence of H1R in CD4+ T cells and its interaction with histamine regulates early TCR signals that lead to Th1 differentiation and autoimmune disease.
Collapse
Affiliation(s)
- Rajkumar Noubade
- Department of Medicine, University of Vermont, Burlington, Vermont, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Yu N, Atienza JM, Bernard J, Blanc S, Zhu J, Wang X, Xu X, Abassi YA. Real-time monitoring of morphological changes in living cells by electronic cell sensor arrays: an approach to study G protein-coupled receptors. Anal Chem 2007; 78:35-43. [PMID: 16383308 DOI: 10.1021/ac051695v] [Citation(s) in RCA: 186] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
G protein-coupled receptors (GPCRs) constitute important targets for drug discovery against a wide range of ailments including cancer, inflammatory, and cardiovascular diseases. Efforts are underway to screen selective modulators of GPCRs and also to deorphanize GPCRs with unidentified natural ligands. Most GPCR-based cellular screens depend on labeling or recombinant expression of receptor or reporter proteins, which may not capture the true physiology or pharmacology of the GPCRs. In this paper, we describe a noninvasive and label-free assay for GPCRs that can be used with both engineered and nonengineered cell lines. The assay is based on using cell-electrode impedance to measure minute changes in cellular morphology as a result of ligand-dependent GPCR activation. We have used this technology to assay the functional activation of GPCRs coupled to different signaling pathways and have compared it to standard assays. We have used pharmacological modulators of GPCR signaling pathways to demonstrate the specificity of impedance-based measurements. Our data indicate that cell-electrode impedance measurements offer a convenient, sensitive, and quantitative method for assessing GPCR function. Moreover, the noninvasive nature of the readout offers the added advantage of performing multiple treatments in the same well to study events such as desensitization and receptor cross-talk.
Collapse
Affiliation(s)
- Naichen Yu
- ACEA Biosciences, 11585 Sorrento Valley Road, San Diego, California 92121, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Das AK, Yoshimura S, Mishima R, Fujimoto K, Mizuguchi H, Dev S, Wakayama Y, Kitamura Y, Horio S, Takeda N, Fukui H. Stimulation of histamine H1 receptor up-regulates histamine H1 receptor itself through activation of receptor gene transcription. J Pharmacol Sci 2007; 103:374-82. [PMID: 17409634 DOI: 10.1254/jphs.fp0061411] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Histamine is a major mediator in allergy acting mainly through the histamine H(1) receptor (H1R). Although H1R up-regulation has been suggested as an important step for induction of allergic symptoms, little is known about the regulation of H1R level. Here we report that the activation of H1R up-regulates H1R through augmentation of H1R mRNA expression in HeLa cells. Histamine stimulation significantly increased both H1R promoter activity and mRNA level without alteration in mRNA stability. H1R protein was also up-regulated by histamine. An H1R antagonist but not histamine H(2) receptor antagonist blocked histamine-induced up-regulation of both promoter activity and mRNA expression. A protein kinase C (PKC) activator, phorbol-12-myristate-13-acetate, increased H1R mRNA expression, whereas an activator of PKA or PKG (8-Br-cAMP or 8-Br-cGMP, respectively) did not. Furthermore, histamine-induced up-regulation of both promoter activity and mRNA level were completely suppressed by the PKC inhibitor Ro-31-8220. H1R antagonists have long been thought to block H1R and inhibit immediate allergy symptoms. In addition to this short-term effect, our data propose their long-term inhibitory effect against allergic diseases by suppressing PKC-mediated H1R gene transcription. This finding provides new insights into the therapeutic target of H1R antagonist in allergic diseases.
Collapse
Affiliation(s)
- Asish K Das
- Department of Molecular Pharmacology, Graduate School of Health Biosciences, The University of Tokushima, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Self TJ, Oakley SM, Hill SJ. Clathrin-independent internalization of the human histamine H1-receptor in CHO-K1 cells. Br J Pharmacol 2006; 146:612-24. [PMID: 16086035 PMCID: PMC1751180 DOI: 10.1038/sj.bjp.0706337] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The aim of the present study was to investigate the cellular pathway involved in histamine-stimulated internalization of the human H1-receptor in CHO-K1 cells expressing N-terminal myc-tagged H1-receptor (Myc-H1) or N-terminal myc-C-terminal green fluorescent protein (Myc-GFP H1) versions of the receptor. Studies of 3H-mepyramine binding and histamine-stimulated 3H-inositol phosphate accumulation in these cells showed that the Myc-H1 and Myc-GFP H1-receptors had identical pharmacology to the wild-type H1-receptor. The Myc-H1-receptor was rapidly internalized in CHO-K1 cells following stimulation with histamine (0.1 mM). This response occurred within 15 min, and could be prevented by the quaternary H1-receptor antagonist alpha-pirdonium. Similar data were obtained with the Myc-GFP H1-receptors. Internalization of the Myc-GFP H1-receptor was maintained in the absence of extracellular calcium and was not inhibited by the CAM kinase II inhibitor KN-62 (10 microM). Phorbol dibutyrate, an activator of protein kinase C, was also able to stimulate internalization of the H1-receptor. However, inhibition or downregulation of protein kinase C (which significantly modified histamine-stimulated inositol phosphate responses) was without effect on the internalization of the H1-receptor stimulated by histamine. Hypertonic sucrose did not prevent histamine-induced internalization of the Myc-GFP H1-receptor, but was able to attenuate internalization of transferrin via clathrin-mediated endocytosis in the same cells. In contrast, preincubation of cells with filipin or nystatin, which disrupts caveolae and lipid rafts, completely inhibited the histamine-induced internalization of the Myc-GFP H1-receptor, but was without effect on the sequestration of transferrin. The H1-receptor and cholera toxin subunit B were colocalized under resting conditions at the cell surface. Immunohistochemical studies with an antibody to caveolin-1 confirmed that this protein was also localized predominantly to the plasma membrane. However, following stimulation of CHO-Myc-GFP H1 cells with histamine, there was no evidence for internalization of caveolin-1 in parallel with the H1-receptor. These data provide strong evidence that the H1-receptor is internalized via a clathrin-independent mechanism and most likely involves lipid rafts.
Collapse
Affiliation(s)
- Timothy J Self
- Institute of Cell Signalling, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH
| | - Sarah M Oakley
- Institute of Cell Signalling, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH
| | - Stephen J Hill
- Institute of Cell Signalling, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH
- Author for correspondence:
| |
Collapse
|
17
|
Kharmate G, Liu Z, Patterson E, Khan MM. Histamine affects STAT6 phosphorylation via its effects on IL-4 secretion: role of H1 receptors in the regulation of IL-4 production. Int Immunopharmacol 2006; 7:277-86. [PMID: 17276885 PMCID: PMC1913482 DOI: 10.1016/j.intimp.2006.10.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2006] [Revised: 10/20/2006] [Accepted: 10/26/2006] [Indexed: 01/29/2023]
Abstract
Signal Transducer and Activator of Transcription (STAT)-6 is a transcriptional factor activated mainly through the cytokines IL-4 and IL-13 leading to the Th2 cell differentiation. Th2 cells play a role in the etiology and pathogenesis of allergic disease. Histamine alters the Th1/Th2 cytokine balance towards the Th2 cytokine profile and consequently plays a role in allergic diseases and asthma. This study was designed to investigate the effects of histamine on the STAT6 phosphorylation. C57/BL6 splenocytes were pretreated with different concentrations of histamine (10(-)(4) M to 10(-)(13) M) followed by stimulation with PMA+ionomycin or IL-4. The phosphorylated and total basal STAT6 levels were assessed by employing the immunoblotting technique. Histamine caused the hyper-phosphorylation of STAT6. H1 receptor antagonist pyrilamine reversed the effect of histamine on STAT6 phosphorylation. However, H2 receptor antagonist ranitidine and H3/H4 receptor antagonist thioperamide did not affect the histamine mediated hyper-phosphorylation of STAT6. Furthermore, H1 receptor agonist betahistine enhanced the phosphorylation of STAT6 whereas H2 receptor agonist amthamine did not affect the phosphorylation STAT6. Furthermore, tyrosine kinase inhibitor, tyrphostin, inhibited the histamine mediated phosphorylation of STAT6 when stimulated with PMA+ionomycin. The effects of histamine on the STAT6 phosphorylation were indirect since they were blocked either by the antibodies to IL-4 and IL-13 or in IL-4 knock out mice in the presence of IL-13 antibody. These observations suggest that histamine indirectly affected the STAT6 phosphorylation via its effects on the secretion of cytokines (IL-4) and H1 receptor played a role in this process.
Collapse
Affiliation(s)
- Geetanjali Kharmate
- Departments of Pharmaceutical Sciences, Creighton University Medical Center, Omaha, NE 68178
| | - Zhongfeng Liu
- Departments of Pharmaceutical Sciences, Creighton University Medical Center, Omaha, NE 68178
| | - Eric Patterson
- Biomedical Sciences, Creighton University Medical Center, Omaha, NE 68178
| | - Manzoor M. Khan
- **Corresponding author: Manzoor M. Khan, Ph.D., Department of Pharmaceutical Sciences, Creighton University, Omaha, NE 68178, Phone: 402-280-5576, Fax: 402-280-1883, E-mail:
| |
Collapse
|
18
|
Matsuyama K, Ichikawa T, Nitta Y, Ikoma Y, Ishimura K, Horio S, Fukui H. Localized expression of histamine H1 receptors in syncytiotrophoblast cells of human placenta. J Pharmacol Sci 2006; 102:331-7. [PMID: 17099293 DOI: 10.1254/jphs.fp0060862] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
The previous Northern blot analysis and in situ hybridization studies showed that histamine H1-receptor (H1R) mRNA is expressed in human placenta and suggested that H(1)R plays some roles in the function of placenta in pregnancy. To investigate further, it is essential to show the precise location of H1R in the placenta. In the present study, we investigated H1R expression in human placenta by radioligand binding assay and immunohistochemical study using an antibody against human H1R. Placentas were obtained from normal uncomplicated deliveries. Membranes prepared from the tissue exhibited saturable [3H]mepyramine binding (K(d) = 4.0 +/- 0.6 nM and B(max) = 91.4 +/- 4.9 fmol/mg of protein). Stereoisomers of chlorpheniramine inhibited [(3)H]mepyramine binding; d-chlorpheniramine inhibited more potently than l-chlorpheniramine, K(i) values being 1.1 +/- 0.4 and 270 +/- 170 nM, respectively. The placenta tissues were positively immunostained with anti-H1R antibody only in the region of the syncytiotrophoblast of chorionic villus. The tissues were double stained with anti-H1R antibody and an antibody against human chorionic gonadotoropin (hCG) that is solely expressed in placental syncytiotrophoblast cells. The results showed that H1R and hCG were expressed on the same cells, that is, syncytiotrophoblast cells. These results indicate that H1Rs are specifically expressed in syncytiotrophoblast cells of human placenta organ.
Collapse
Affiliation(s)
- Kazuto Matsuyama
- Department of Molecular Pharmacology, Institute of Health and Biosciences, The University of Tokushima, Tokushima, Japan
| | | | | | | | | | | | | |
Collapse
|
19
|
Lipnik-Stangelj M. Multiple role of histamine H1-receptor-PKC-MAPK signalling pathway in histamine-stimulated nerve growth factor synthesis and secretion. Biochem Pharmacol 2006; 72:1375-81. [PMID: 16884695 DOI: 10.1016/j.bcp.2006.06.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2006] [Revised: 06/16/2006] [Accepted: 06/21/2006] [Indexed: 11/20/2022]
Abstract
Histamine is a potent stimulator of nerve growth factor (NGF) production in the central nerve system and in the periphery as well. In this review, the biochemical mechanisms of histamine-stimulated NGF synthesis and secretion, and interactions between histamine, interleukin-1beta, and interleukin-6 are discussed. The main signalling pathway, involved in the stimulation of NGF production by histamine, includes activation of histamine H(1)-receptor, stimulation of Ca(2+)-dependent protein kinase C and mitogen-activated protein kinase. The same signalling pathway is involved in the interactions between histamine, interleukin-1beta, and interleukin-6, where NGF secretion is amplified. Whereas histamine and interleukin-1beta cause additive stimulatory effect on NGF secretion, interaction between histamine and interleukin-6 causes a long-term synergism. Thus, activation of histamine H(1)-receptor-protein kinase C-mitogen-activated protein kinase signalling pathway plays a crucial role not only in the direct stimulation of NGF secretion by histamine, but also in the indirect stimulation via different types of interactions between histamine, interleukin-1beta, and interleukin-6, which may have important therapeutic implications in modulation of NGF production.
Collapse
Affiliation(s)
- Metoda Lipnik-Stangelj
- Department of Pharmacology and Experimental Toxicology, Faculty of Medicine, Korytkova 2, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
20
|
Liu Z, Kharmate G, Patterson E, Khan MM. Role of H1 receptors in histamine-mediated up-regulation of STAT4 phosphorylation. Int Immunopharmacol 2006; 6:485-93. [PMID: 16428084 DOI: 10.1016/j.intimp.2005.09.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2005] [Revised: 04/11/2005] [Accepted: 09/23/2005] [Indexed: 11/19/2022]
Abstract
Histamine shifts TH1/TH2 cytokine balance from TH1 to TH2 cytokines and regulates the function of lymphocytes after binding to histamine receptors. The phosphorylation of STAT factors and the translocation to the nucleus are important steps in the regulation of TH1/TH2 cytokine balance. This study was designed to investigate the effects of histamine on the phosphorylation of STAT4. C57BL/6 splenocytes were isolated and treated with histamine (10(-4) to 10(-9) M) after activation with either PMA (phorbol 12 myristate 13-acetate) plus ionomycin or IL-12. The phosphorylated STAT4 levels were analyzed by Western Blot Analysis. Unstimulated splenocytes expressed both STAT4 and phosphorylated STAT4. However, phosphorylated STAT4 gradually declined within 24 h. Histamine increased the phosphorylation of STAT4 at lower concentrations (10(-6) to 10(-9) M), and had no effect at higher concentrations (10(-4) and 10(-5) M) after the cells were stimulated with PMA + ionomycin. Histamine did not affect IL-12-induced phosphorylation of STAT4. To characterize the histamine receptor subtypes involved in the up-regulation of STAT4 phosphorylation, various H1, H2 and H3/H4 receptor antagonists and/or agonists were employed. H1 receptor agonist (betahistine), but not H2 receptor agonist (amthamine), induced phosphorylation of STAT4. H1 receptor antagonist (pyrilamine) inhibited histamine-mediated phosphorylation of STAT4. However, H2 receptor antagonist (ranitidine) and H3/H4 receptor antagonist (thioperamide) did not alter this effect. Tyrosine kinase inhibitor (tyrphostin) failed to block histamine-mediated phosphorylation of STAT4. These observations suggest that histamine up-regulated the phosphorylation of STAT4 via H1 receptors, and that the Ca2+-PKC pathway, but not the tyrosine kinase pathway, was involved in this effect.
Collapse
Affiliation(s)
- Zhongfeng Liu
- Department of Pharmaceutical Sciences, Creighton University Medical Center, Omaha, NE 68178, USA
| | | | | | | |
Collapse
|
21
|
Maruko T, Nakahara T, Sakamoto K, Saito M, Sugimoto N, Takuwa Y, Ishii K. Involvement of the βγ subunits of G proteins in the cAMP response induced by stimulation of the histamine H1 receptor. Naunyn Schmiedebergs Arch Pharmacol 2005; 372:153-9. [PMID: 16189696 DOI: 10.1007/s00210-005-0001-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2005] [Accepted: 08/17/2005] [Indexed: 11/29/2022]
Abstract
Stimulation of the histamine H1 receptor has been shown to enhance adenosine 3', 5'-cyclic monophosphate (cAMP) accumulation in various cell types but, to date, the mechanism by which this occurs is still unclear. In the present study, we examined the possibility that the betagamma subunits of G proteins (G betagamma) are involved in this process in cultured Chinese hamster ovary cells transfected with the human histamine H1 receptor (CHO-H1). Histamine increased intracellular cAMP levels in a concentration-dependent manner in CHO-H1 cells, and this histamine action was abolished by pyrilamine (1 microM). Inhibition of histamine H1 receptor-G(q) protein coupling by stable expression of the C-terminal peptide of G alpha(q) protein significantly attenuated the cAMP accumulation induced by histamine. By comparison, neither BAPTA/AM (50 microM), an intracellular Ca2+ chelator, nor GF 109203X (1 microM), an inhibitor of protein kinase C, influenced the cAMP response. Histamine H1 receptor-mediated cAMP accumulation was significantly inhibited by transient transfection of CHO-H1 cells with the C-terminal peptide of beta-adrenoceptor kinase I (residues 542-685), a scavenger of G betagamma. Stable expression of the C-terminal peptide of the G alpha(s) protein, but not treatment with pertussis toxin (200 ng/ml for 24 h), attenuated the histamine H1 receptor-mediated cAMP accumulation. These results suggest that stimulation of histamine H1 receptors activates adenylyl cyclase through the release of G betagamma subunits from G proteins, thereby elevating intracellular cAMP levels.
Collapse
Affiliation(s)
- Takeshi Maruko
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | | | | | | | | | | | | |
Collapse
|
22
|
Matsubara M, Tamura T, Ohmori K, Hasegawa K. Histamine H1 receptor antagonist blocks histamine-induced proinflammatory cytokine production through inhibition of Ca2+-dependent protein kinase C, Raf/MEK/ERK and IKK/IκB/NF-κB signal cascades. Biochem Pharmacol 2005; 69:433-49. [PMID: 15652235 DOI: 10.1016/j.bcp.2004.10.006] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2004] [Accepted: 10/20/2004] [Indexed: 11/23/2022]
Abstract
Histamine H1 receptor (H1R), a therapeutic target for alleviation of acute allergic reaction, may be also involved in mediating inflammatory responses via effects on cytokine production. However, the mechanisms whereby histamine induces cytokine production are poorly defined. In this study, we comprehensively investigated the signaling pathway involved in cytokine expression caused by histamine, using native human epidermal keratinocytes. We confirmed the expression of functional H1R by reverse transcription-polymerase chain reaction (RT-PCR), Western blotting and histamine-induced Ca(2+) elevation. Histamine induced concentration- and time-dependent production of granulocyte-macrophage-colony stimulating factor (GM-CSF), interleukin (IL)-8 and IL-6, which was completely blocked by olopatadine, an H1 antagonist. Histamine activated the phosphorylation of protein kinase C (PKC), c-Raf, mitogen-activated protein/extracellular signal-regulated kinase kinase (MEK), extracellular signal-regulated kinase (ERK), I kappa B kinase (IKK), inhibitory kappa B (I kappa B)-alpha and nuclear factor-KB (NF-kappa B) p65, which was inhibited by Ro-31-8220, a PKC inhibitor. Also, Ro-31-8220 significantly suppressed the expression of these cytokines. BAPTA-AM, an intracellular Ca(2+) chelator, also reduced PKC phosphorylation and cytokine expression. PD98059, a MEK inhibitor, and BAY 11-8702, an I kappa B-alpha inhibitor, reduced ERK and NF-kappa B cascade activation, respectively, with little effect on PKC phosphorylation. PD98059 preferentially inhibited GM-CSF production whereas BAY 11-8702 prevented IL-8 and IL-6 production. Furthermore, in addition to the above cytokines, histamine stimulated the biosynthesis and/or release of numerous keratinocyte-derived mediators, which are probably regulated by the ERK or NF-kappa B cascades. Our study suggests that histamine activates Ca(2+)-dependent PKC isoforms that play crucial roles in the activation of Raf/MEK/ERK and IKK/I kappa B/NF-kappa B cascades, leading to up-regulation of cytokine expression. Thus, the anti-inflammatory benefit of H1 antagonists may be in part due to prevention of cytokine production.
Collapse
Affiliation(s)
- Masahiro Matsubara
- Department of Pharmacology and Molecular Biology, Pharmaceutical Research Center, Kyowa Hakko Kogyo Co. Ltd., Shizuoka 411-8731, Japan.
| | | | | | | |
Collapse
|
23
|
Galeotti N, Malmberg-Aiello P, Bartolini A, Schunack W, Ghelardini C. H1-receptor stimulation induces hyperalgesia through activation of the phospholipase C-PKC pathway. Neuropharmacology 2004; 47:295-303. [PMID: 15223308 DOI: 10.1016/j.neuropharm.2004.03.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2004] [Revised: 02/24/2004] [Accepted: 03/18/2004] [Indexed: 10/26/2022]
Abstract
The supraspinal cellular events involved in H(1)-mediated hyperalgesia were investigated in a condition of acute thermal pain by means of the mouse hot-plate test. I.c.v. administration of the phospholipase C (PLC) inhibitors U-73122 and neomycin antagonized the hyperalgesia induced by the selective H(1) agonist FMPH. By contrast, U-73343, an analogue of U-73122 used as negative control, was unable to modify the reduction of the pain threshold induced by FMPH. In mice undergoing treatment with LiCl, which impairs phosphatidylinositol synthesis, or treatment with heparin, an IP(3)-receptor antagonist, the hyperalgesia induced by the H(1)-receptor agonist remained unchanged. Similarly, pretreatment with D-myo inositol did not alter the H(1)-induced hypernociceptive response. Neither i.c.v. pretreatment with TMB-8, a blocker of Ca(2+) release from intracellular stores, nor pretreatment with thapsigargin, a depletor of Ca(2+) intracellular stores, prevented the decrease of pain threshold induced by FMPH. On the other hand, i.c.v. pretreatment with the selective protein kinase C (PKC) inhibitors calphostin C and chelerytrine resulted in a dose-dependent prevention of the H(1)-receptor agonist-induced hyperalgesia. The administration of PKC activators, such as PMA and PDBu, did not produce any effect on FMPH effect. The pharmacological treatments employed did not produce any behavioral impairment of mice as revealed by the rota-rod and hole-board tests. These results indicate a role for the PLC-PKC pathway in central H(1)-induced hyperalgesia in mice. Furthermore, activation of PLC-IP(3) did not appear to play a major role in the modulation of pain perception by H(1)-receptor agonists.
Collapse
Affiliation(s)
- Nicoletta Galeotti
- Department of Preclinical and Clinical Pharmacology, Viale G. Pieraccini 6, I-50139 Florence, Italy.
| | | | | | | | | |
Collapse
|
24
|
Kanda N, Watanabe S. Histamine enhances the production of granulocyte-macrophage colony-stimulating factor via protein kinase Calpha and extracellular signal-regulated kinase in human keratinocytes. J Invest Dermatol 2004; 122:863-72. [PMID: 15102074 DOI: 10.1111/j.0022-202x.2004.22432.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The production of granulocyte-macrophage colony-stimulating factor (GM-CSF) in keratinocytes is related to the chronicity of atopic dermatitis. Mast cell-derived histamine contributes to the cross-talk between mast cells and keratinocytes. We examined the effects of histamine on GM-CSF production in human keratinocytes. Histamine increased GM-CSF secretion, mRNA stability and promoter activity. Activator protein-1 (AP-1) and nuclear factor-kappaB (NF-kappaB) elements on the promoter were responsible for the activation by histamine. Histamine enhanced transcriptional activity and DNA binding of AP-1 and NF-kappaB. Histamine shifted AP-1 composition form c-Jun homodimers to c-Fos/c-Jun heterodimers, and transiently expressed c-Fos protein. Histamine rapidly induced the phosphorylation and degradation of inhibitory kappaB. Histamine induced membrane translocation of protein kinase Calpha. Histamine-induced GM-CSF production was completely abolished by H1 antagonist pyrilamine and conventional protein kinase C inhibitor Gö6976, and partially suppressed by PD98059 which inhibits the activation of extracellular signal-regulated kinase. Gö6976 and PD98059 suppressed histamine-induced c-Fos expression and AP-1 activation. Gö6976 and PD98059 suppressed histamine-induced enhancement of NF-kappaB transcriptional activity. Histamine-induced phosphorylation and degradation of inhibitory kappaB was suppressed by Gö6976, but not by PD98059. These results suggest that histamine may enhance GM-CSF production at transcriptional and posttranscriptional levels via H1 receptor, protein kinase Calpha and extracellular signal-regulated kinase.
Collapse
Affiliation(s)
- Naoko Kanda
- Department of Dermatology, Teikyo University, School of Medicine, Tokyo, Japan.
| | | |
Collapse
|
25
|
Fitzsimons CP, Monczor F, Fernández N, Shayo C, Davio C. Mepyramine, a histamine H1 receptor inverse agonist, binds preferentially to a G protein-coupled form of the receptor and sequesters G protein. J Biol Chem 2004; 279:34431-9. [PMID: 15192105 DOI: 10.1074/jbc.m400738200] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Accurate characterization of the molecular mechanisms of the action of ligands is an extremely important issue for their appropriate research, pharmacological, and therapeutic uses. In view of this fact, the aim of the present work was to investigate the mechanisms involved in the actions of mepyramine at the guinea pig H(1) receptor stably expressed in Chinese hamster ovary cells. We found that mepyramine is able to decrease the basal constitutive activity of the guinea pig H(1) receptor, to bind with high affinity to a G(q/11) protein-coupled form of the receptor and to promote a G protein-coupled inactive state of the H(1) receptor that interferes with the G(q/11)-mediated signaling of the endogenously expressed ATP receptor, as predicted by the Cubic Ternary Complex Model of receptor occupancy. The effect of mepyramine on ATP-induced signaling was specifically neutralized by Galpha(11) overexpression, indicating that mepyramine is able to reduce G protein availability for other non-related receptors associated with the same signaling pathway. Finally, we found a loss of mepyramine efficacy in decreasing basal levels of intracellular calcium at high Galpha(11) expression levels, which can be theoretically explained in terms of high H(1) receptor constitutive activity. The whole of the present work sheds new light on H(1) receptor pharmacology and the mechanisms H(1) receptor inverse agonists could use to exert their observed negative efficacy.
Collapse
Affiliation(s)
- Carlos P Fitzsimons
- Laboratorio de Radioisótopos, Facultad de Farmacia y Bioquímica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires 1113, Argentina
| | | | | | | | | |
Collapse
|
26
|
Baker JG, Hall IP, Hill SJ. Temporal characteristics of cAMP response element-mediated gene transcription: requirement for sustained cAMP production. Mol Pharmacol 2004; 65:986-98. [PMID: 15044629 DOI: 10.1124/mol.65.4.986] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Many clinically used drugs are G-protein-coupled receptor (GPCR) antagonists and are given long-term to prevent receptor activation by endogenous agonists. Most GPCR antagonists are considered to have little agonist efficacy of their own. However, many beta antagonists do stimulate very small beta(2) adrenoceptor-mediated cAMP responses, but these responses become substantial at the level of cAMP response element (CRE)-gene transcription. Here, we compared the temporal characteristics of these beta(2) adrenoceptor-mediated cAMP and CRE-gene transcription responses with ligands of differing agonist efficacy. Within a minute, full agonists (e.g., isoprenaline) stimulated large increases in intracellular and exported cAMP. Very weak partial agonists (e.g., alprenolol) did not increase intracellular cAMP (only stimulating a small export). However, all agonists (regardless of efficacy) stimulated an increase in CRE-gene transcription after a 2-h incubation. An initial 30-min continual stimulation was required to initiate the process of CRE-gene transcription for all ligands. Longer agonist incubations resulted in larger gene transcription responses in a proportional manner for both weak and full agonists alike, and this was despite the lack of intracellular cAMP detection for the weaker ligands. Thus, the major initiator for CRE-gene transcription was not cAMP concentration or total quantity generated but a sustained turnover of intracellular cAMP and hence sustained stimulation of CREB phosphorylation. Thus, long-acting agonists and long-term treatments with very weak partial agonists (including many drugs classified previously as antagonists based on traditional second-messenger assays, e.g., several clinically used "beta-blockers") may cause more substantial gene transcription than previously believed.
Collapse
Affiliation(s)
- Jillian G Baker
- Institute of Cell Signaling, Queen's Medical Centre, Nottingham NG7 2UH, United Kingdom.
| | | | | |
Collapse
|
27
|
Baker JG, Hall IP, Hill SJ. Agonist and inverse agonist actions of beta-blockers at the human beta 2-adrenoceptor provide evidence for agonist-directed signaling. Mol Pharmacol 2004; 64:1357-69. [PMID: 14645666 DOI: 10.1124/mol.64.6.1357] [Citation(s) in RCA: 156] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Beta-blockers have beneficial effects in heart failure, although the underlying mechanism is unknown. Beta2-adrenoceptors, however, are proportionally higher in the failing human heart. This study shows several clinically used beta-blockers are agonists at the human beta2-adrenoceptor. Although these agonist effects were small at the cAMP level, they were substantial at the level of cAMP response element (CRE)-mediated gene transcription. Some of the effects of "beta-blockers" seen in heart failure may be related to the beta2-agonist actions of these compounds. CRE-gene transcription responses to beta2-agonists, forskolin, and cAMP-analogs were sensitive to p42/44-mitogen-activated protein (MAP) kinase pathway inhibitors. p42/44-MAP kinase activation was also shown directly by western blotting and enzyme-linked immunosorbent assay techniques. N-[2-(4-bromocinnamylamino)ethyl]-5-isoquinoline (H89; a protein kinase A inhibitor) stimulated cAMP accumulation and CRE gene transcription via the beta2-adrenoceptor at concentrations at which protein kinase A was inhibited, providing evidence for an alternative pathway. Propranolol, however, produced paradoxical effects; it reduced basal cAMP accumulation (via beta2-mediated inverse agonism) but stimulated beta2-mediated CRE gene transcription. This cannot be explained by a sequential pathway from Gs-adenylyl cyclase-cAMP to CRE binding protein phosphorylation. Both responses to propranolol were insensitive to pertussis toxin, thus excluding Gi-protein involvement. Propranolol CRE gene transcription responses were attenuated by p42/44-MAP kinase inhibitors and propranolol was also found to directly stimulate the p42/44-MAP kinase pathway. Studies of inositol phosphate accumulation and of protein kinase C or Rho kinase inhibitors on CRE-gene transcription provided no evidence for Gq/11 or G12/13 involvement. These data suggest that propranolol can simultaneously act as an inverse agonist through a Gs-coupled mechanism while stimulating the p42/44-MAP kinase pathway through an alternative G-protein-independent mechanism.
Collapse
Affiliation(s)
- Jillian G Baker
- Institute of Cell Signaling, Queen's Medical Centre, Nottingham NG7 2UH, UK.
| | | | | |
Collapse
|
28
|
Greenaway EC, Sepulveda MF, Cunningham FM, Goode NT. Protein kinase C (PKC) isotype profile in eosinophils from ponies with sweet itch and role in histamine-induced eosinophil activation. Vet Immunol Immunopathol 2003; 96:53-63. [PMID: 14522134 DOI: 10.1016/s0165-2427(03)00161-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Eosinophils have been implicated in the pathogenesis of the seasonal equine allergic skin disease, sweet itch. Protein kinase C (PKC) is involved in regulating eosinophil function and antigen challenge has been reported to alter PKC isotype expression in blood eosinophils from allergic human subjects. Here we have compared the pattern of PKC isotype expression in eosinophils from sweet itch ponies with that in cells from normal ponies both during the active and inactive phases of the disease. A role for PKC in histamine-induced eosinophil activation was also investigated. Conventional PKCs alpha and beta, novel PKCs delta and epsilon and atypical PKCs iota and zeta were identified in eosinophils pooled from four allergic ponies during the inactive phase, when no clinical signs were evident. The PKC isotypes, like those in eosinophils from normal ponies, were located primarily in the particulate fraction of the cell. Isotype expression in cells from normal and allergic animals did not appear to be different. In contrast, during the active phase of the disease, when the sweet itch ponies had clinical signs, the expression of PKCs beta, epsilon and iota in eosinophils from these animals appeared to be increased relative to that in cells from normal ponies. When PKC expression in eosinophils from five individual normal and sweet itch ponies was compared, small, but statistically significant, increases in PKC epsilon and PKCdelta expression were evident in eosinophils from the sweet itch ponies during the active and inactive phases, respectively. The non-selective PKC inhibitors, staurosporine and Ro31-8220, significantly reduced histamine-induced superoxide production. Use of Gö6976, an inhibitor of conventional PKCs, suggested that PKCalpha and/or beta were involved and that there was significantly greater inhibition of the response in eosinophils obtained from sweet itch ponies during the active phase. There was no significant difference in histamine-induced superoxide production by eosinophils from allergic and normal ponies and the functional significance of the increased PKC isotype expression in eosinophils from sweet itch ponies relative to that in cells from healthy animals remains to be established.
Collapse
Affiliation(s)
- E C Greenaway
- Department of Veterinary Basic Sciences, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hertfordshire AL9 7TA, UK
| | | | | | | |
Collapse
|
29
|
Kanda N, Watanabe S. Histamine enhances the production of nerve growth factor in human keratinocytes. J Invest Dermatol 2003; 121:570-7. [PMID: 12925217 DOI: 10.1046/j.1523-1747.2003.12428.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Nerve growth factor induces innervation and epidermal hyperplasia in inflammatory skin diseases like psoriasis. Nerve growth factor production by keratinocytes is increased in the inflammatory lesions. Nerve growth factor induces histamine release from mast cells. We examined the in vitro effects of histamine on nerve growth factor production in human keratinocytes. Histamine enhanced nerve growth factor secretion, mRNA expression, and promoter activity in keratinocytes. Two TPA-response elements on the nerve growth factor promoter were responsible for the activation by histamine. Histamine enhanced transcriptional activity and DNA binding of activator protein 1 at the two TPA-response elements. It shifted the TPA-response-element-binding activator protein 1 composition from c-Jun homodimers to c-Fos/c-Jun heterodimers. Histamine transiently induced c-Fos mRNA expression, which was not detectable in unstimulated keratinocytes, whereas c-Jun mRNA expression was constitutive and was not altered by histamine. Histamine-induced enhancement of nerve growth factor secretion, promoter activity, activator protein 1 transcriptional activity, and c-Fos expression was suppressed by H1 antagonist pyrilamine, protein kinase C inhibitor calphostin C, and PD98059, an inhibitor of mitogen-activated protein kinase kinase 1. Histamine induced the translocation of protein kinase C activity from cytosol to membrane, which was suppressed by phospholipase C inhibitor U73122. It stimulated the phosphorylation of extracellular signal-regulated kinase, which was blocked by pyrilamine, calphostin C, and PD98059. These results suggest that histamine may enhance nerve growth factor production by inducing c-Fos expression in keratinocytes. These effects may be mediated by the H1-receptor-induced signaling cascade of phospholipase C-protein kinase C-mitogen-activated protein kinase kinase 1-extracellular signal-regulated kinase.
Collapse
Affiliation(s)
- Naoko Kanda
- Department of Dermatology, Teikyo University, School of Medicine, 11-1 Kaga-2, Itabashi-Ku, Tokyo 173-8605, Japan.
| | | |
Collapse
|
30
|
Hill KJ, Webber AC, Hill SJ. A role of protein kinase C mu in signalling from the human adenosine A1 receptor to the nucleus. Br J Pharmacol 2003; 139:721-32. [PMID: 12812995 PMCID: PMC1573890 DOI: 10.1038/sj.bjp.0705294] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2002] [Revised: 02/05/2003] [Accepted: 03/25/2003] [Indexed: 11/09/2022] Open
Abstract
1 Stimulation of adenosine A(1) receptors produced a stimulation of c-fos promoter-regulated gene transcription in Chinese hamster ovary (CHO)-A1 cells expressing the human A(1) receptor. Gene transcription was monitored using a luciferase-based reporter gene (pGL3). 2 This response to the A(1) receptor agonist N(6)-cyclopentyladenosine (CPA) was sensitive to inhibition by pertussis toxin, the MEK-1 inhibitor PD 98059 and by the phosphatidylinositol-3-kinase inhibitors wortmannin and LY 294002. The response was also completely abolished by the protein kinase C (PKC) inhibitor Ro-31-8220. 3 Several isoforms of PKC can be detected in CHO-A1 cells (alpha, delta, epsilon, micro, iota, zeta), but only PKC alpha, PKC delta and PKC were downregulated by prolonged treatment with phorbol ester. The c-fos-regulated luciferase response to A(1) agonists was not, however, inhibited by 24 h pretreatment with the phorbol esters phorbol 12,13-dibutyrate (PDBu). This observation, together with the fact that a significant attenuation (40%) of the c-fos-luciferase response to PDBu and A(1) agonist was produced by low concentrations of the PKC inhibitor Gö 6976 suggests a role for PKC micro. 4 Stimulation of CHO-A1 cells with CPA stimulated the activation of endogenous PKC micro as measured by autophosphorylation. This was rapid, occurred within 1-2 min, but returned to basal levels after 30 min. Furthermore, transient expression of a constitutively active form of PKC micro resulted in a significant increase in c-fos-regulated gene expression. 5 Taken together, these data suggest that PKC micro plays an important role in the ability of the adenosine A(1) receptor to signal to the nucleus.
Collapse
Affiliation(s)
- Kathryn J Hill
- Institute of Cell Signalling, Medical School, Queen's Medical Centre, Nottingham NG7 2UH
| | - Anne C Webber
- Institute of Cell Signalling, Medical School, Queen's Medical Centre, Nottingham NG7 2UH
| | - Stephen J Hill
- Institute of Cell Signalling, Medical School, Queen's Medical Centre, Nottingham NG7 2UH
| |
Collapse
|
31
|
Berg KA, Evans KLJ, Cropper JD, Clarke WP. Temporal regulation of agonist efficacy at 5-hydroxytryptamine (5-HT)1A and 5-HT 1B receptors. J Pharmacol Exp Ther 2003; 304:200-5. [PMID: 12490592 DOI: 10.1124/jpet.102.042564] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Coactivation of purinergic (P 2Y) receptors reduces agonist efficacy at serotonin 1B (5-HT 1B), but not 5-HT 1A receptors. Herein, we report that pretreatment for 5 min with the P 2Y receptor agonist ATP reduced agonist responsiveness at the 5-HT 1A, but not at the 5-HT 1B, receptor. The effect of ATP pretreatment on the 5-HT 1A receptor response rapidly reversed within a 10 min time frame between P 2Y receptor and 5-HT 1A receptor activation. ATP pretreatment effects on 5-HT 1A agonist responsiveness were blocked by the protein kinase inhibitors staurosporine and bisindolylmaleimide, suggesting that the ATP-mediated temporal regulation involves activation of protein kinase C (PKC). Moreover, the temporal effect of ATP was blocked by incubation with 1% ethanol, suggesting that consequences of phospholipase D (PLD) activation play a role. ATP pretreatment blocked the inhibitory effect produced by 5-HT 2C receptor activation on the 5-HT 1A, but not the 5-HT 1B, receptor response, suggesting that the 5-HT 1A receptor itself was the target for PLD/PKC action. Finally, ethanol did not block the reduction in responsiveness of the 5-HT 1A receptor system produced by activation of PKC with phorbol ester treatment, suggesting that PKC activation lies downstream of PLD. Taken together, these data suggest that activation of P 2Y receptors can reduce responsiveness of the 5-HT 1A receptor system via a PLD/PKC-dependent mechanism that is highly dependent upon the temporal pattern of receptor activation. Moreover, this work underscores the importance of time as a variable in receptor signaling cross talk and serves to further illustrate differences between the 5-HT 1A and 5-HT 1B receptor systems.
Collapse
Affiliation(s)
- Kelly A Berg
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, Texas 78229-3900, USA.
| | | | | | | |
Collapse
|
32
|
Dickenson JM. Stimulation of protein kinase B and p70 S6 kinase by the histamine H1 receptor in DDT1MF-2 smooth muscle cells. Br J Pharmacol 2002; 135:1967-76. [PMID: 11959800 PMCID: PMC1573327 DOI: 10.1038/sj.bjp.0704664] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2001] [Revised: 12/04/2001] [Accepted: 02/01/2002] [Indexed: 12/19/2022] Open
Abstract
1. Previous studies have shown that the histamine H(1) receptor activates p42/p44 mitogen-activated protein kinases (MAPK) in DDT(1)MF-2 smooth muscle cells via a phosphatidylinositol 3-kinase (PI-3K)-dependent pathway. In this study the effect of histamine H(1) receptor stimulation on protein kinase B (PKB) and p70 S6 kinase, both of which are downstream targets of PI-3K, has been investigated. Increases in PKB and p70 S6 kinase activation were monitored by Western blotting using phospho-specific PKB (Ser(473)) and p70 S6 kinase (Thr(421)/Ser(424)) antibodies. 2. Histamine stimulated time and concentration-dependent increases in the phosphorylation of PKB and p70 S6 kinase in DDT(1)MF-2 cells. Both responses were completely inhibited by the histamine H(1) receptor antagonist mepyramine and following pre-treatment with pertussis toxin, to block G(i)/G(o) protein dependent pathways. 3. The PI-3K inhibitors wortmannin (IC(50) 5.9+/-0.5 nM) and LY 294002 (IC(50) 6.9+/-0.8 microM) attenuated the increase in PKB phosphorylation induced by histamine (100 microM) in a concentration-dependent manner. 4. Histamine-induced increases in p70 S6 kinase phosphorylation were partially sensitive to rapamycin (20 nM; 68% inhibition) but completely blocked by wortmannin (100 nM), LY 294002 (30 microM) and the MAPK kinase inhibitor PD 98059 (50 microM). 5. In summary, these data demonstrate that the histamine H(1) receptor stimulates PKB and p70 S6 kinase phosphorylation in DDT(1)MF-2 smooth muscle cells. However, functional studies revealed that histamine does not stimulate DDT(1)MF-2 cell proliferation or attenuate staurosporine-induced caspase-3 activity. The challenge for future research will be to link the stimulation of these kinase pathways with the physiological and pathophysiological roles of the histamine H(1) receptor.
Collapse
Affiliation(s)
- John M Dickenson
- Department of Life Sciences, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK.
| |
Collapse
|