1
|
Howell A, Arsic N, Griebel P. Resting and activated bovine neutrophils and eosinophils differ in their responses to adrenergic agonists. Vet Immunol Immunopathol 2024; 272:110758. [PMID: 38669937 DOI: 10.1016/j.vetimm.2024.110758] [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: 01/17/2024] [Revised: 04/09/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024]
Abstract
Polymorphonuclear cells (PMN) provide a rapid response to infection and tissue damage and stress can modify these critical innate immune defences. The study of adrenergic receptor (AR) expression and function in bovine PMNs is limited but both neutrophils and eosinophils express numerous AR genes but differ significantly in their expression of individual AR genes. A flow cytometric technique was developed to differentiate between bovine neutrophils and eosinophils so both neutrophil and eosinophil responses to adrenergic agonists could be analysed. Neutrophils and eosinophils displayed significantly different changes in CD11b, L-selectin, and CD44 expression when activated by bovine serum opsonized zymosan and recombinant bovine interferon gamma. The responses of activated and resting neutrophils and eosinophils were then compared following stimulation with endogenous adrenergic agonists, epinephrine (E) norepinephrine (NE), and synthetic agonists targeting α1-, α2-, or β-ARs. Both resting and activated neutrophils and eosinophils displayed differences in iROS, CD44, and L-selectin expression following stimulation with E and NE. Resting neutrophils displayed pro-inflammatory responses to both E and NE, while resting eosinophils displayed a pro-inflammatory response to only NE. No single synthetic adrenergic agonist fully recapitulated responses observed with either E or NE and responses to adrenergic agonists were dose-dependent. In conclusion, bovine eosinophils and neutrophils responded to multiple adrenergic agonists by altering expression of proteins involved in immune surveillance and pro-inflammatory responses. Significant differences in neutrophil and eosinophil responses to adrenergic agonists are consistent with their differences in AR gene expression. This highlights the importance of analysing separately these two PMN subpopulations when investigating the effects of either endogenous or synthetic AR agonists.
Collapse
Affiliation(s)
- Angela Howell
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, 120-Veterinary Road, Saskatoon, SK S7N 5E3, Canada; Global Institute for Food Security (GIFS), University of Saskatchewan, 421 Downey Rd, Saskatoon, SK S7N 4L8, Canada
| | - Natasa Arsic
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, 120-Veterinary Road, Saskatoon, SK S7N 5E3, Canada
| | - Philip Griebel
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, 120-Veterinary Road, Saskatoon, SK S7N 5E3, Canada; School of Public Health, 104 Clinic Place, University of Saskatchewan, SK S7N 2Z4, Canada.
| |
Collapse
|
2
|
DelaCuesta-Barrutia J, Hidema S, Caldwell HK, Nishimori K, Erdozain AM, Peñagarikano O. In need of a specific antibody against the oxytocin receptor for neuropsychiatric research: A KO validation study. J Psychiatr Res 2024; 173:260-270. [PMID: 38554622 DOI: 10.1016/j.jpsychires.2024.03.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 02/22/2024] [Accepted: 03/21/2024] [Indexed: 04/02/2024]
Abstract
Antibodies are one of the most utilized tools in biomedical research. However, few of them are rigorously evaluated, as there are no accepted guidelines or standardized methods for determining their validity before commercialization. Often, an antibody is considered validated if it detects a band by Western blot of the expected molecular weight and, in some cases, if blocking peptides result in loss of staining. Neither of these approaches are unquestionable proof of target specificity. Since the oxytocin receptor has recently become a popular target in neuropsychiatric research, the need for specific antibodies to be used in brain has arisen. In this work, we have tested the specificity of six commercially available oxytocin receptor antibodies, indicated by the manufacturers to be suitable for Western blot and with an available image showing the correct size band (45-55 KDa). Antibodies were first tested by Western blot in brain lysates of wild-type and oxytocin receptor knockout mice. Uterus tissue was also tested as control for putative differential tissue specificity. In brain, the six tested antibodies lacked target specificity, as both wild-type and receptor knockout samples resulted in a similar staining pattern, including the expected 45-55 KDa band. Five of the six antibodies detected a selective band in uterus (which disappeared in knockout tissue). These five specific antibodies were also tested for immunohistochemistry in uterus, where only one was specific. However, when the uterine-specific antibody was tested in brain tissue, it lacked specificity. In conclusion, none of the six tested commercial antibodies are suitable to detect oxytocin receptor in brain by either Western blot or immunohistochemistry, although some do specifically detect it in uterus. The present work highlights the need to develop standardized antibody validation methods, including a proper negative control, in order to grant quality and reproducibility of the generated data.
Collapse
Affiliation(s)
- Jon DelaCuesta-Barrutia
- Department of Pharmacology, School of Medicine, University of the Basque Country (UPV/EHU), Leioa, 48940, Spain
| | - Shizu Hidema
- Department of Obesity and Inflammation, Fukushima Medical University, Fukushima, 960-1295, Japan
| | - Heather K Caldwell
- Department of Biological Sciences and School of Biomedical Sciences, Kent State University, Ohio, 44242, USA
| | - Katsuhiko Nishimori
- Department of Obesity and Inflammation, Fukushima Medical University, Fukushima, 960-1295, Japan
| | - Amaia M Erdozain
- Department of Pharmacology, School of Medicine, University of the Basque Country (UPV/EHU), Leioa, 48940, Spain; Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Leioa, 48940, Spain
| | - Olga Peñagarikano
- Department of Pharmacology, School of Medicine, University of the Basque Country (UPV/EHU), Leioa, 48940, Spain; Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Leioa, 48940, Spain.
| |
Collapse
|
3
|
Abdul-Ridha A, de Zhang LA, Betrie AH, Deluigi M, Vaid TM, Whitehead A, Zhang Y, Davis B, Harris R, Simmonite H, Hubbard RE, Gooley PR, Plückthun A, Bathgate RA, Chalmers DK, Scott DJ. Identification of a Novel Subtype-Selective α 1B-Adrenoceptor Antagonist. ACS Chem Neurosci 2024; 15:671-684. [PMID: 38238043 PMCID: PMC10854767 DOI: 10.1021/acschemneuro.3c00767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 02/08/2024] Open
Abstract
α1A-, α1B-, and α1D-adrenoceptors (α1-ARs) are members of the adrenoceptor G protein-coupled receptor family that are activated by adrenaline (epinephrine) and noradrenaline. α1-ARs are clinically targeted using antagonists that have minimal subtype selectivity, such as prazosin and tamsulosin, to treat hypertension and benign prostatic hyperplasia, respectively. Abundant expression of α1-ARs in the heart and central nervous system (CNS) makes these receptors potential targets for the treatment of cardiovascular and CNS disorders, such as heart failure, epilepsy, and Alzheimer's disease. Our understanding of the precise physiological roles of α1-ARs, however, and their involvement in disease has been hindered by the lack of sufficiently subtype-selective tool compounds, especially for α1B-AR. Here, we report the discovery of 4-[(2-hydroxyethyl)amino]-6-methyl-2H-chromen-2-one (Cpd1), as an α1B-AR antagonist that has 10-15-fold selectivity over α1A-AR and α1D-AR. Through computational and site-directed mutagenesis studies, we have identified the binding site of Cpd1 in α1B-AR and propose the molecular basis of α1B-AR selectivity, where the nonconserved V19745.52 residue plays a major role, with contributions from L3146.55 within the α1B-AR pocket. By exploring the structure-activity relationships of Cpd1 at α1B-AR, we have also identified 3-[(cyclohexylamino)methyl]-6-methylquinolin-2(1H)-one (Cpd24), which has a stronger binding affinity than Cpd1, albeit with reduced selectivity for α1B-AR. Cpd1 and Cpd24 represent potential leads for α1B-AR-selective drug discovery and novel tool molecules to further study the physiology of α1-ARs.
Collapse
Affiliation(s)
- Alaa Abdul-Ridha
- The
Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
| | - Lazarus A. de Zhang
- The
Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
| | | | - Mattia Deluigi
- Department
of Biochemistry, University of Zurich, CH-8057 Zurich, Switzerland
| | - Tasneem M. Vaid
- The
Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
- The
Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria 3010, Australia
- The Bio21
Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Alice Whitehead
- The
Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
| | - Yifan Zhang
- The
Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
| | - Ben Davis
- Vernalis
(R&D) Ltd, Granta Park, Cambridge CB21 6GB, U.K.
| | - Richard Harris
- Vernalis
(R&D) Ltd, Granta Park, Cambridge CB21 6GB, U.K.
| | | | - Roderick E. Hubbard
- Vernalis
(R&D) Ltd, Granta Park, Cambridge CB21 6GB, U.K.
- Department
of Chemistry, University of York, York YO10 5DD, U.K.
| | - Paul R. Gooley
- The
Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria 3010, Australia
- The Bio21
Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Andreas Plückthun
- Department
of Biochemistry, University of Zurich, CH-8057 Zurich, Switzerland
| | - Ross A.D. Bathgate
- The
Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
- The
Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - David K. Chalmers
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Daniel J. Scott
- The
Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
- The
Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
4
|
Zhang J, Sandroni PB, Huang W, Gao X, Oswalt L, Schroder MA, Lee S, Shih YYI, Huang HYS, Swigart PM, Myagmar BE, Simpson PC, Rossi JS, Schisler JC, Jensen BC. Cardiomyocyte Alpha-1A Adrenergic Receptors Mitigate Postinfarct Remodeling and Mortality by Constraining Necroptosis. JACC Basic Transl Sci 2024; 9:78-96. [PMID: 38362342 PMCID: PMC10864988 DOI: 10.1016/j.jacbts.2023.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 08/26/2023] [Accepted: 08/29/2023] [Indexed: 02/17/2024]
Abstract
Clinical studies have shown that α1-adrenergic receptor antagonists (α-blockers) are associated with increased heart failure risk. The mechanism underlying that hazard and whether it arises from direct inhibition of cardiomyocyte α1-ARs or from systemic effects remain unclear. To address these issues, we created a mouse with cardiomyocyte-specific deletion of the α1A-AR subtype and found that it experienced 70% mortality within 7 days of myocardial infarction driven, in part, by excessive activation of necroptosis. We also found that patients taking α-blockers at our center were at increased risk of death after myocardial infarction, providing clinical correlation for our translational animal models.
Collapse
Affiliation(s)
- Jiandong Zhang
- Division of Cardiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Peyton B. Sandroni
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Wei Huang
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Xiaohua Gao
- Department of Epidemiology, University of North Carolina Gillings School of Public Health, Chapel Hill, North Carolina, USA
| | - Leah Oswalt
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Melissa A. Schroder
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - SungHo Lee
- Center for Animal MRI, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Yen-Yu I. Shih
- Center for Animal MRI, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Hsiao-Ying S. Huang
- Mechanical and Aerospace Engineering Department, North Carolina State University, Raleigh, North Carolina, USA
| | - Philip M. Swigart
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
- San Francisco VA Medical Center, San Francisco, California, USA
| | - Bat E. Myagmar
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
- San Francisco VA Medical Center, San Francisco, California, USA
| | - Paul C. Simpson
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
- San Francisco VA Medical Center, San Francisco, California, USA
| | - Joseph S. Rossi
- Division of Cardiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Jonathan C. Schisler
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Brian C. Jensen
- Division of Cardiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- UNC McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
5
|
Ruan Y, Buonfiglio F, Gericke A. Adrenoceptors in the Eye - Physiological and Pathophysiological Relevance. Handb Exp Pharmacol 2024; 285:453-505. [PMID: 38082203 DOI: 10.1007/164_2023_702] [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] [Indexed: 09/05/2024]
Abstract
The autonomic nervous system plays a crucial role in the innervation of the eye. Consequently, it comes as no surprise that catecholamines and their corresponding receptors have been extensively studied and characterized in numerous ocular structures, including the cornea, conjunctiva, lacrimal gland, trabecular meshwork, uvea, and retina. These investigations have unveiled substantial clinical implications, particularly in the context of treating glaucoma, a progressive neurodegenerative disorder responsible for irreversible vision loss on a global scale. The primary therapeutic approaches for glaucoma frequently involve the modulation of α1-, α2-, and β-adrenoceptors, making them pivotal targets. In this chapter, we offer a comprehensive overview of the expression, distribution, and functional roles of adrenoceptors within various components of the eye and its associated structures. Additionally, we delve into the pivotal role of adrenoceptors in the pathophysiology of glaucoma. Furthermore, we provide a concise historical perspective on adrenoceptor research, examine the distinct contributions of individual adrenoceptor subtypes to the treatment of various ocular conditions, and propose potential future avenues of exploration in this field.
Collapse
Affiliation(s)
- Yue Ruan
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Francesco Buonfiglio
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
6
|
Ge M, Wu L, He F, Tai Y, Fang R, Han D, Guo P, Liu H, Hu Y, Xu S, Wei W, Wang Q. CP-25 inhibits the hyperactivation of rheumatic synoviocytes by suppressing the switch in G αs-G αi coupling to the β 2-adrenergic receptor. Cell Commun Signal 2023; 21:346. [PMID: 38037039 PMCID: PMC10688045 DOI: 10.1186/s12964-023-01358-z] [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: 07/17/2023] [Accepted: 10/16/2023] [Indexed: 12/02/2023] Open
Abstract
In essence, the β2 adrenergic receptor (β2AR) plays an antiproliferative role by increasing the intracellular cyclic 3',5'-adenosine monophosphate (cAMP) concentration through Gαs coupling, but interestingly, β2AR antagonists are able to effectively inhibit fibroblast-like synoviocytes (FLSs) proliferation, thus ameliorating experimental RA, indicating that the β2AR signalling pathway is impaired in RA FLSs via unknown mechanisms. The local epinephrine (Epi) level was found to be much higher in inflammatory joints than in normal joints, and high-level stimulation with Epi or isoproterenol (ISO) directly promoted FLSs proliferation and migration due to impaired β2AR signalling and cAMP production. By applying inhibitor of receptor internalization, and small interfering RNA (siRNA) of Gαs and Gαi, and by using fluorescence resonance energy transfer and coimmunoprecipitation assays, a switch in Gαs-Gαi coupling to β2AR was observed in inflammatory FLSs as well as in FLSs with chronic ISO stimulation. This Gαi coupling was then revealed to be initiated by G protein coupled receptor kinase 2 (GRK2) but not β-arrestin2 or protein kinase A-mediated phosphorylation of β2AR. Inhibiting the activity of GRK2 with the novel GRK2 inhibitor paeoniflorin-6'-O-benzene sulfonate (CP-25), a derivative of paeoniflorin, or the accepted GRK2 inhibitor paroxetine effectively reversed the switch in Gαs-Gαi coupling to β2AR during inflammation and restored the intracellular cAMP level in ISO-stimulated FLSs. As expected, CP-25 significantly inhibited the hyperplasia of FLSs in a collagen-induced arthritis (CIA) model (CIA FLSs) and normal FLSs stimulated with ISO and finally ameliorated CIA in rats. Together, our findings revealed the pathological changes in β2AR signalling in CIA FLSs, determined the underlying mechanisms and identified the pharmacological target of the GRK2 inhibitor CP-25 in treating CIA. Video Abstract.
Collapse
Affiliation(s)
- Mingli Ge
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, 230032, China
| | - Li Wu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, 230032, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Feng He
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, 230032, China
| | - Yu Tai
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, 230032, China
| | - Ruhong Fang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, 230032, China
| | - Dafei Han
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, 230032, China
| | - Paipai Guo
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, 230032, China
| | - Hao Liu
- School of Pharmacy, Bengbu Medical College, Bengbu, 233030, China
| | - Yong Hu
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Shenglin Xu
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China.
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, 230032, China.
| | - Qingtong Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, 230032, China.
| |
Collapse
|
7
|
Thibault RT, Amaral OB, Argolo F, Bandrowski AE, Davidson AR, Drude NI. Open Science 2.0: Towards a truly collaborative research ecosystem. PLoS Biol 2023; 21:e3002362. [PMID: 37856538 PMCID: PMC10617723 DOI: 10.1371/journal.pbio.3002362] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 10/31/2023] [Indexed: 10/21/2023] Open
Abstract
Conversations about open science have reached the mainstream, yet many open science practices such as data sharing remain uncommon. Our efforts towards openness therefore need to increase in scale and aim for a more ambitious target. We need an ecosystem not only where research outputs are openly shared but also in which transparency permeates the research process from the start and lends itself to more rigorous and collaborative research. To support this vision, this Essay provides an overview of a selection of open science initiatives from the past 2 decades, focusing on methods transparency, scholarly communication, team science, and research culture, and speculates about what the future of open science could look like. It then draws on these examples to provide recommendations for how funders, institutions, journals, regulators, and other stakeholders can create an environment that is ripe for improvement.
Collapse
Affiliation(s)
- Robert T. Thibault
- 1 Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California, Unites States of America
| | - Olavo B. Amaral
- Institute of Medical Biochemistry Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Anita E. Bandrowski
- FAIR Data Informatics Lab, Department of Neuroscience, UCSD, San Diego, California, United States of America
- SciCrunch Inc., San Diego, California, United States of America
| | - Alexandra R, Davidson
- Institute for Evidence-Based Health Care, Bond University, Robina, Australia
- Faculty of Health Science and Medicine, Bond University, Robina, Australia
| | - Natascha I. Drude
- Berlin Institute of Health (BIH) at Charité, BIH QUEST Center for Responsible Research, Berlin, Germany
| |
Collapse
|
8
|
Pérez-Santos I, García-Cabezas MÁ, Cavada C. Mapping the primate thalamus: systematic approach to analyze the distribution of subcortical neuromodulatory afferents. Brain Struct Funct 2023:10.1007/s00429-023-02619-w. [PMID: 36890350 DOI: 10.1007/s00429-023-02619-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 02/09/2023] [Indexed: 03/10/2023]
Abstract
Neuromodulatory afferents to thalamic nuclei are key for information transmission and thus play critical roles in sensory, motor, and limbic processes. Over the course of the last decades, diverse attempts have been made to map and describe subcortical neuromodulatory afferents to the primate thalamus, including axons using acetylcholine, serotonin, dopamine, noradrenaline, adrenaline, and histamine. Our group has been actively involved in this endeavor. The published descriptions on neuromodulatory afferents to the primate thalamus have been made in different laboratories and are not fully comparable due to methodological divergences (for example, fixation procedures, planes of cutting, techniques used to detect the afferents, different criteria for identification of thalamic nuclei…). Such variation affects the results obtained. Therefore, systematic methodological and analytical approaches are much needed. The present article proposes reproducible methodological and terminological frameworks for primate thalamic mapping. We suggest the use of standard stereotaxic planes to produce and present maps of the primate thalamus, as well as the use of the Anglo-American school terminology (vs. the German school terminology) for identification of thalamic nuclei. Finally, a public repository of the data collected under agreed-on frameworks would be a useful tool for looking up and comparing data on the structure and connections of primate thalamic nuclei. Important and agreed-on efforts are required to create, manage, and fund a unified and homogeneous resource of data on the primate thalamus. Likewise, a firm commitment of the institutions to preserve experimental brain material is much needed because neuroscience work with non-human primates is becoming increasingly rare, making earlier material still more valuable.
Collapse
Affiliation(s)
- Isabel Pérez-Santos
- Department of Anatomy, Histology and Neuroscience, School of Medicine, Facultad de Medicina, Universidad Autónoma de Madrid, Calle Arzobispo Morcillo 4, 28029, Madrid, Spain.,PhD Program in Neuroscience, Universidad Autónoma de Madrid-Cajal, Madrid, Spain
| | - Miguel Ángel García-Cabezas
- Department of Anatomy, Histology and Neuroscience, School of Medicine, Facultad de Medicina, Universidad Autónoma de Madrid, Calle Arzobispo Morcillo 4, 28029, Madrid, Spain.,PhD Program in Neuroscience, Universidad Autónoma de Madrid-Cajal, Madrid, Spain.,Neural Systems Laboratory, Department of Health Sciences, Boston University, Boston, MA, USA
| | - Carmen Cavada
- Department of Anatomy, Histology and Neuroscience, School of Medicine, Facultad de Medicina, Universidad Autónoma de Madrid, Calle Arzobispo Morcillo 4, 28029, Madrid, Spain. .,PhD Program in Neuroscience, Universidad Autónoma de Madrid-Cajal, Madrid, Spain.
| |
Collapse
|
9
|
Perez DM. α 1-Adrenergic Receptors: Insights into Potential Therapeutic Opportunities for COVID-19, Heart Failure, and Alzheimer's Disease. Int J Mol Sci 2023; 24:4188. [PMID: 36835598 PMCID: PMC9963459 DOI: 10.3390/ijms24044188] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/22/2023] Open
Abstract
α1-Adrenergic receptors (ARs) are members of the G-Protein Coupled Receptor superfamily and with other related receptors (β and α2), they are involved in regulating the sympathetic nervous system through binding and activation by norepinephrine and epinephrine. Traditionally, α1-AR antagonists were first used as anti-hypertensives, as α1-AR activation increases vasoconstriction, but they are not a first-line use at present. The current usage of α1-AR antagonists increases urinary flow in benign prostatic hyperplasia. α1-AR agonists are used in septic shock, but the increased blood pressure response limits use for other conditions. However, with the advent of genetic-based animal models of the subtypes, drug design of highly selective ligands, scientists have discovered potentially newer uses for both agonists and antagonists of the α1-AR. In this review, we highlight newer treatment potential for α1A-AR agonists (heart failure, ischemia, and Alzheimer's disease) and non-selective α1-AR antagonists (COVID-19/SARS, Parkinson's disease, and posttraumatic stress disorder). While the studies reviewed here are still preclinical in cell lines and rodent disease models or have undergone initial clinical trials, potential therapeutics discussed here should not be used for non-approved conditions.
Collapse
Affiliation(s)
- Dianne M Perez
- The Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH 44195, USA
| |
Collapse
|
10
|
Vinit S, Michel-Flutot P, Mansart A, Fayssoil A. Effects of C2 hemisection on respiratory and cardiovascular functions in rats. Neural Regen Res 2023; 18:428-433. [PMID: 35900441 PMCID: PMC9396504 DOI: 10.4103/1673-5374.346469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
High cervical spinal cord injuries induce permanent neuromotor and autonomic deficits. These injuries impact both central respiratory and cardiovascular functions through modulation of the sympathetic nervous system. So far, cardiovascular studies have focused on models of complete contusion or transection at the lower cervical and thoracic levels and diaphragm activity evaluations using invasive methods. The present study aimed to evaluate the impact of C2 hemisection on different parameters representing vital functions (i.e., respiratory function, cardiovascular, and renal filtration parameters) at the moment of injury and 7 days post-injury in rats. No ventilatory parameters evaluated by plethysmography were impacted during quiet breathing after 7 days post-injury, whereas permanent diaphragm hemiplegia was observed by ultrasound and confirmed by diaphragmatic electromyography in anesthetized rats. Interestingly, the mean arterial pressure was reduced immediately after C2 hemisection, with complete compensation at 7 days post-injury. Renal filtration was unaffected at 7 days post-injury; however, remnant systolic dysfunction characterized by a reduced left ventricular ejection fraction persisted at 7 days post-injury. Taken together, these results demonstrated that following C2 hemisection, diaphragm activity and systolic function are impacted up to 7 days post-injury, whereas the respiratory and cardiovascular systems display vast adaptation to maintain ventilatory parameters and blood pressure homeostasis, with the latter likely sustained by the remaining descending sympathetic inputs spared by the initial injury. A better broad characterization of the physiopathology of high cervical spinal cord injuries covering a longer time period post-injury could be beneficial for understanding evaluations of putative therapeutics to further increase cardiorespiratory recovery.
Collapse
|
11
|
Bomholt AB, Johansen CD, Christensen JB, Kjeldsen SAS, Galsgaard KD, Winther-Sørensen M, Serizawa R, Hornum M, Porrini E, Pedersen J, Ørskov C, Gluud LL, Sørensen CM, Holst JJ, Albrechtsen R, Wewer Albrechtsen NJ. Evaluation of commercially available glucagon receptor antibodies and glucagon receptor expression. Commun Biol 2022; 5:1278. [PMID: 36418521 PMCID: PMC9684523 DOI: 10.1038/s42003-022-04242-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2022] Open
Abstract
Glucagon is a major regulator of metabolism and drugs targeting the glucagon receptor (GCGR) are being developed. Insight into tissue and cell-specific expression of the GCGR is important to understand the biology of glucagon and to differentiate between direct and indirect actions of glucagon. However, it has been challenging to localize the GCGR in tissue due to low expression levels and lack of specific methods. Immunohistochemistry has frequently been used for GCGR localization, but antibodies targeting G-protein-coupled-receptors may be inaccurate. We evaluated all currently commercially available GCGR antibodies. The antibody, ab75240 (Antibody no. 11) was found to perform best among the twelve antibodies tested and using this antibody we found expression of the GCGR in the kidney, liver, preadipocytes, pancreas, and heart. Three antibody-independent approaches all confirmed the presence of the GCGR within the pancreas, liver and the kidneys. GCGR expression should be evaluated by both antibody and antibody-independent approaches.
Collapse
Affiliation(s)
- Anna Billeschou Bomholt
- grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christian Dall Johansen
- grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark ,grid.5254.60000 0001 0674 042XNovo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Bager Christensen
- grid.5254.60000 0001 0674 042XDepartment of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sasha Alexandra Sampson Kjeldsen
- grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark ,grid.5254.60000 0001 0674 042XNovo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Katrine Douglas Galsgaard
- grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark ,grid.5254.60000 0001 0674 042XNovo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marie Winther-Sørensen
- grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark ,grid.5254.60000 0001 0674 042XNovo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Reza Serizawa
- grid.4973.90000 0004 0646 7373Department of Pathology, Copenhagen University Hospital, Hvidovre, Denmark
| | - Mads Hornum
- grid.475435.4Department of Nephrology, Centre for Cancer and Organ Diseases, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark ,grid.5254.60000 0001 0674 042XDepartment of Clinical Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Esteban Porrini
- grid.411220.40000 0000 9826 9219Instituto de Tecnologías Biomédicas, University of La Laguna, Research Unit, Hospital Universitario de Canarias, Tenerife, Spain
| | - Jens Pedersen
- grid.5254.60000 0001 0674 042XDepartment of Clinical Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark ,grid.411900.d0000 0004 0646 8325Department of Internal Medicine, Endocrinology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Cathrine Ørskov
- grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lise Lotte Gluud
- grid.5254.60000 0001 0674 042XDepartment of Clinical Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark ,grid.4973.90000 0004 0646 7373Gastro Unit, Copenhagen University Hospital, Hvidovre, Denmark
| | - Charlotte Mehlin Sørensen
- grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark ,grid.5254.60000 0001 0674 042XNovo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Reidar Albrechtsen
- grid.5254.60000 0001 0674 042XBiotech Research & Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Nicolai Jacob Wewer Albrechtsen
- grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark ,grid.5254.60000 0001 0674 042XNovo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark ,grid.512917.9Department of Clinical Biochemistry, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| |
Collapse
|
12
|
Deeney BT, Cao G, Orfanos S, Lee J, Kan M, Himes BE, Parikh V, Koziol-White CJ, An SS, Panettieri RA. Epinephrine evokes shortening of human airway smooth muscle cells following β 2 adrenergic receptor desensitization. Am J Physiol Lung Cell Mol Physiol 2022; 323:L142-L151. [PMID: 35787178 PMCID: PMC9359643 DOI: 10.1152/ajplung.00444.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 06/14/2022] [Accepted: 06/28/2022] [Indexed: 11/22/2022] Open
Abstract
Epinephrine (EPI), an endogenous catecholamine involved in the body's fight-or-flight responses to stress, activates α1-adrenergic receptors (α1ARs) expressed on various organs to evoke a wide range of physiological functions, including vasoconstriction. In the smooth muscle of human bronchi, however, the functional role of EPI on α1ARs remains controversial. Classically, evidence suggests that EPI promotes bronchodilation by stimulating β2-adrenergic receptors (β2ARs). Conventionally, the selective β2AR agonism of EPI was thought to be, in part, due to a predominance of β2ARs and/or a sparse, or lack of α1AR activity in human airway smooth muscle (HASM) cells. Surprisingly, we find that HASM cells express a high abundance of ADRA1B (the α1AR subtype B) and identify a spontaneous "switch-like" activation of α1ARs that evokes intracellular calcium, myosin light chain phosphorylation, and HASM cell shortening. The switch-like responses, and related EPI-induced biochemical and mechanical signals, emerged upon pharmacological inhibition of β2ARs and/or under experimental conditions that induce β2AR tachyphylaxis. EPI-induced procontractile effects were abrogated by an α1AR antagonist, doxazosin mesylate (DM). These data collectively uncover a previously unrecognized feed-forward mechanism driving bronchospasm via two distinct classes of G protein-coupled receptors (GPCRs) and provide a basis for reexamining α1AR inhibition for the management of stress/exercise-induced asthma and/or β2-agonist insensitivity in patients with difficult-to-control, disease subtypes.
Collapse
Affiliation(s)
- Brian T Deeney
- Rutgers Institute for Translational Medicine and Science, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Gaoyuan Cao
- Rutgers Institute for Translational Medicine and Science, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Sarah Orfanos
- Rutgers Institute for Translational Medicine and Science, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Jordan Lee
- The Joint Graduate Program in Toxicology, Department of Pharmacology and Toxicology, Rutgers-Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Mengyuan Kan
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Blanca E Himes
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Vishal Parikh
- Rutgers Institute for Translational Medicine and Science, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Cynthia J Koziol-White
- Rutgers Institute for Translational Medicine and Science, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Steven S An
- Rutgers Institute for Translational Medicine and Science, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
- The Joint Graduate Program in Toxicology, Department of Pharmacology and Toxicology, Rutgers-Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| |
Collapse
|
13
|
Lehtimäki J, Jalava N, Unkila K, Aspegren J, Haapalinna A, Pesonen U. Tasipimidine-the pharmacological profile of a novel orally active selective α 2A-adrenoceptor agonist. Eur J Pharmacol 2022; 923:174949. [PMID: 35405115 DOI: 10.1016/j.ejphar.2022.174949] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/05/2022] [Indexed: 11/03/2022]
Abstract
The pharmacological profile of tasipimidine, a novel orally active α2-adrenoceptor agonist developed for situational anxiety and fear in dogs, was studied in various in vitro and in vivo models. In the cell assays, tasipimidine demonstrated binding affinity and full agonism on the human α2A-adrenoceptors with a pEC50 of 7.57, while agonism on the α2B-and α2C-adrenoceptors and the rodent α2D-adrenoceptor was weaker, resulting in pEC50 values of 6.00, 6.29 and 6.56, respectively. Tasipimidine had a low binding affinity on the human α1-adrenoceptors. It had no functional effects in the LNCaP cells expressing endogenously the human α1A-adrenoceptors but was a weak agonist in the Chem-1 cells coexpressing Gα15 protein and α1A-adrenoceptors. In the recombinant CHO cells, although tasipimidine was a weak partial agonist in the inositol monophosphate accumulation assay, it was a full agonist in the intracellular [Ca2+] assay. No functional effects were observed on the human α1B-adrenoceptor, whereas in the rat α1A and α1B-adrenoceptors, tasipimidine was a weak partial agonist. In the rat vas deferens preparations, tasipimidine was a full agonist on the α2D-adrenoceptor but weak partial agonist on the α1-adrenoceptor. The receptor profile of tasipimidine indicated few secondary targets, and no functional effects were observed. Sedative effects of tasipimidine were demonstrated in vivo by the reduced acoustic startle reflex in rats with subcutaneous doses and decreased spontaneous locomotor activity in mice with subcutaneous and higher oral doses. It may be concluded that tasipimidine is an orally active and selective α2A-adrenoceptor agonist.
Collapse
Affiliation(s)
- Jyrki Lehtimäki
- Orion Corporation Orion Pharma, R&D, P.O. Box 425, FI-20101, Turku, Finland.
| | - Niina Jalava
- Orion Corporation Orion Pharma, R&D, P.O. Box 425, FI-20101, Turku, Finland
| | - Kaisa Unkila
- Orion Corporation Orion Pharma, R&D, P.O. Box 425, FI-20101, Turku, Finland
| | - John Aspegren
- Orion Corporation Orion Pharma, R&D, P.O. Box 425, FI-20101, Turku, Finland
| | - Antti Haapalinna
- Orion Corporation Orion Pharma, R&D, P.O. Box 425, FI-20101, Turku, Finland
| | - Ullamari Pesonen
- Institute of Biomedicine, Faculty of Medicine, University of Turku, Kiinamyllynkatu 10, FI-20520, Turku, Finland
| |
Collapse
|
14
|
Gq neuromodulation of BLA parvalbumin interneurons induces burst firing and mediates fear-associated network and behavioral state transition in mice. Nat Commun 2022; 13:1290. [PMID: 35277502 PMCID: PMC8917207 DOI: 10.1038/s41467-022-28928-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 02/14/2022] [Indexed: 01/08/2023] Open
Abstract
Patterned coordination of network activity in the basolateral amygdala (BLA) is important for fear expression. Neuromodulatory systems play an essential role in regulating changes between behavioral states, however the mechanisms underlying this neuromodulatory control of transitions between brain and behavioral states remain largely unknown. We show that chemogenetic Gq activation and α1 adrenoreceptor activation in mouse BLA parvalbumin (PV) interneurons induces a previously undescribed, stereotyped phasic bursting in PV neurons and time-locked synchronized bursts of inhibitory postsynaptic currents and phasic firing in BLA principal neurons. This Gq-coupled receptor activation in PV neurons suppresses gamma oscillations in vivo and in an ex vivo slice model, and facilitates fear memory recall, which is consistent with BLA gamma suppression during conditioned fear expression. Thus, here we identify a neuromodulatory mechanism in PV inhibitory interneurons of the BLA which regulates BLA network oscillations and fear memory recall. The authors study mechanisms underlying neuromodulatory control of transitions between brain and behavioral states. They identify a mechanism whereby modulation of Gq activity in basolateral amygdala parvalbumin interneurons mediates the transition to a fear-associated network and behavioral state.
Collapse
|
15
|
Klussmeier A, Aurich S, Niederstadt L, Wiedenmann B, Grötzinger C. Secretin Receptor as a Target in Gastrointestinal Cancer: Expression Analysis and Ligand Development. Biomedicines 2022; 10:biomedicines10030536. [PMID: 35327338 PMCID: PMC8944975 DOI: 10.3390/biomedicines10030536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 02/01/2023] Open
Abstract
Secretin was originally discovered as a gastrointestinal peptide that stimulates fluid secretion from the pancreas and liver and delays gastric emptying. In disease, a secretin receptor (SCTR) was found to occur as a splice variant in gastrinoma and pancreatic adenocarcinoma. Overexpression of SCTR has been described for gastrinomas, carcinoid tumors of the lung and cholangiocarcinoma. SCTR therefore is considered a candidate target for molecular tumor imaging as well as for peptide receptor radioligand therapy (PRRT) in a number of oncological indications. The aim of this study was to characterize SCTR expression in esophageal and pancreatic cancer, demonstrating for the first time high SCTR overexpression in these tumor types. In total, 65 of 70 pancreatic ductal adenocarcinoma tissues stained strongly positive for SCTR in immunohistochemistry, as did most of the 151 esophageal cancer samples, with minor influence of grading in both entities. In addition, the aim of this study was to further delineate residues in human secretin that are critical for binding to and activation of human SCTR. For a potential development of short and metabolically stable analogs for clinical use, it was intended to probe the peptide for its capacity to incorporate deletions and substitutions without losing its affinity to SCTR. In a systematic approach, a library of 146 secretin variants containing single amino acid substitutions as well as truncations on either end was tested in β-arrestin2-GFP translocation and fluorescent ligand internalization assays employing high-content analysis, in cAMP assays which run in agonist and antagonist mode, and in radioligand binding. The main structural determinants of SCTR binding and activation were localized to the N-terminus, with His1, Asp3 being among the most sensitive positions, followed by Phe6, Thr7 and Leu10. Aminoterminal truncation caused a rapid decline in receptor activity and most of these variants proved to be partial agonists showing antagonistic properties. In this study, the most potent novel antagonist showed an IC50 of 309 ± 74 nM in the β-arrestin2-GFP translocation assay on human SCTR while remaining a weak partial agonist. Future studies will have to demonstrate the utility of further enhanced secretin analogues as tracers for in vivo imaging and therapy.
Collapse
Affiliation(s)
- Anja Klussmeier
- Department of Hepatology and Gastroenterology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (A.K.); (S.A.); (L.N.); (B.W.)
- Institut für Chemie und Biochemie, Fachbereich Biologie, Chemie, Pharmazie, Freie Universität Berlin, 14195 Berlin, Germany
| | - Stefan Aurich
- Department of Hepatology and Gastroenterology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (A.K.); (S.A.); (L.N.); (B.W.)
| | - Lars Niederstadt
- Department of Hepatology and Gastroenterology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (A.K.); (S.A.); (L.N.); (B.W.)
| | - Bertram Wiedenmann
- Department of Hepatology and Gastroenterology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (A.K.); (S.A.); (L.N.); (B.W.)
| | - Carsten Grötzinger
- Department of Hepatology and Gastroenterology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (A.K.); (S.A.); (L.N.); (B.W.)
- Partner Site Berlin, German Cancer Consortium (DKTK), 13353 Berlin, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Correspondence:
| |
Collapse
|
16
|
Howell A, Arsic N, Brownlie R, Griebel P. Adrenergic receptor gene expression in bovine leukocytes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 127:104271. [PMID: 34600023 DOI: 10.1016/j.dci.2021.104271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 06/13/2023]
Abstract
The α- and β-adrenergic receptors (ARs) bind the stress hormones epinephrine (E), norepinephrine (NE), and dopamine and activate diverse physiological responses. A lack of information on AR gene expression in leukocytes limits our understanding of how stress alters immune function. Quantitative analyses of AR gene expression was completed for bovine leukocytes. Individual leukocyte lineages and subpopulations within lineages were isolated with high-speed cell sorting to facilitate a targeted analysis of AR gene expression. These analyses confirmed all 9 AR genes were expressed in bovine leukocytes with marked differences in AR gene expression when comparing among leukocyte lineages. Furthermore, separation of polymorphonuclear cells into neutrophils and eosinophils revealed these key innate immune cells also differ significantly in AR gene expression. This study provides the first comprehensive survey of AR gene expression in immune cells of any mammalian species and provides insight into conflicting reports that stress can either activate or suppress immune function.
Collapse
Affiliation(s)
- Angela Howell
- VIDO-InterVac, University of Saskatchewan, 120-Veterinary Road, Saskatoon, SK, S7K 3J8, Canada
| | - Natasa Arsic
- VIDO-InterVac, University of Saskatchewan, 120-Veterinary Road, Saskatoon, SK, S7K 3J8, Canada
| | - Robert Brownlie
- VIDO-InterVac, University of Saskatchewan, 120-Veterinary Road, Saskatoon, SK, S7K 3J8, Canada
| | - Philip Griebel
- VIDO-InterVac, University of Saskatchewan, 120-Veterinary Road, Saskatoon, SK, S7K 3J8, Canada; School of Public Health, 104 Clinic Place, University of Saskatchewan, SK, S7N 2Z4, Canada.
| |
Collapse
|
17
|
Hoff FW, Horton TM, Kornblau SM. Reverse phase protein arrays in acute leukemia: investigative and methodological challenges. Expert Rev Proteomics 2021; 18:1087-1097. [PMID: 34965151 PMCID: PMC9148717 DOI: 10.1080/14789450.2021.2020655] [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: 09/15/2021] [Accepted: 12/16/2021] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Acute leukemia results from a series of mutational events that alter cell growth and proliferation. Mutations result in protein changes that orchestrate growth alterations characteristic of leukemia. Proteomics is a methodology appropriate for study of protein changes found in leukemia. The high-throughput reverse phase protein array (RPPA) technology is particularly well-suited for the assessment of protein changes in samples derived from clinical trials. AREAS COVERED This review discusses the technical, methodological, and analytical issues related to the successful development of acute leukemia RPPAs. EXPERT COMMENTARY To obtain representative protein sample lysates, samples should be prepared from freshly collected blood or bone marrow material. Variables such as sample shipment, transit time, and holding temperature only have minimal effects on protein expression. CellSave preservation tubes are preferred for cells collected after exposure to chemotherapy, and incorporation of standardized guidelines for antibody validation is recommended. A more systematic biological approach to analyze protein expression is desired, searching for recurrent patterns of protein expression that allow classification of patients into risk groups, or groups of patients that may be treated similarly. Comparing RPPA protein analysis between cell lines and primary samples shows that cell lines are not representative of patient proteomic patterns.
Collapse
Affiliation(s)
- Fieke W. Hoff
- Department of Internal Medicine, UT Southwestern Medical Center, TX, USA
| | - Terzah M. Horton
- Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Steven M. Kornblau
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
18
|
Abstract
Bitter taste-sensing type 2 receptors (TAS2Rs or T2Rs), belonging to the subgroup of family A G-protein coupled receptors (GPCRs), are of crucial importance in the perception of bitterness. Although in the first instance, TAS2Rs were considered to be exclusively distributed in the apical microvilli of taste bud cells, numerous studies have detected these sensory receptor proteins in several extra-oral tissues, such as in pancreatic or ovarian tissues, as well as in their corresponding malignancies. Critical points of extra-oral TAS2Rs biology, such as their structure, roles, signaling transduction pathways, extensive mutational polymorphism, and molecular evolution, have been currently broadly studied. The TAS2R cascade, for instance, has been recently considered to be a pivotal modulator of a number of (patho)physiological processes, including adipogenesis or carcinogenesis. The latest advances in taste receptor biology further raise the possibility of utilizing TAS2Rs as a therapeutic target or as an informative index to predict treatment responses in various disorders. Thus, the focus of this review is to provide an update on the expression and molecular basis of TAS2Rs functions in distinct extra-oral tissues in health and disease. We shall also discuss the therapeutic potential of novel TAS2Rs targets, which are appealing due to their ligand selectivity, expression pattern, or pharmacological profiles.
Collapse
Affiliation(s)
- Kamila Tuzim
- Department of Clinical Pathomorphology, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090, Lublin, Poland.
| | - Agnieszka Korolczuk
- Department of Clinical Pathomorphology, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090, Lublin, Poland
| |
Collapse
|
19
|
Bob-Manuel J, Gautron L. Detection of G Protein-coupled Receptor Expression in Mouse Vagal Afferent Neurons using Multiplex In Situ Hybridization. J Vis Exp 2021:10.3791/62945. [PMID: 34605820 PMCID: PMC9235148 DOI: 10.3791/62945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
This study describes a protocol for the multiplex in situ hybridization (ISH) of the mouse jugular-nodose ganglia, with a particular emphasis on detecting the expression of G protein-coupled receptors (GPCRs). Formalin-fixed jugular-nodose ganglia were processed with the RNAscope technology to simultaneously detect the expression of two representative GPCRs (cholecystokinin and ghrelin receptors) in combination with one marker gene of either nodose (paired-like homeobox 2b, Phox2b) or jugular afferent neurons (PR domain zinc finger protein 12, Prdm12). Labeled ganglia were imaged using confocal microscopy to determine the distribution and expression patterns of the aforementioned transcripts. Briefly, Phox2b afferent neurons were found to abundantly express the cholecystokinin receptor (Cck1r) but not the ghrelin receptor (Ghsr). A small subset of Prdm12 afferent neurons was also found to express Ghsr and/or Cck1r. Potential technical caveats in the design, processing, and interpretation of multiplex ISH are discussed. The approach described in this article may help scientists in generating accurate maps of the transcriptional profiles of vagal afferent neurons.
Collapse
Affiliation(s)
- Johnson Bob-Manuel
- Center for Hypothalamic Research and Department of Internal Medicine, UTSouthwestern Medical Center at Dallas
| | - Laurent Gautron
- Center for Hypothalamic Research and Department of Internal Medicine, UTSouthwestern Medical Center at Dallas;
| |
Collapse
|
20
|
Pérez-Santos I, Palomero-Gallagher N, Zilles K, Cavada C. Distribution of the Noradrenaline Innervation and Adrenoceptors in the Macaque Monkey Thalamus. Cereb Cortex 2021; 31:4115-4139. [PMID: 34003210 PMCID: PMC8328208 DOI: 10.1093/cercor/bhab073] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/18/2021] [Accepted: 03/03/2021] [Indexed: 11/14/2022] Open
Abstract
Noradrenaline (NA) in the thalamus has important roles in physiological, pharmacological, and pathological neuromodulation. In this work, a complete characterization of NA axons and Alpha adrenoceptors distributions is provided. NA axons, revealed by immunohistochemistry against the synthesizing enzyme and the NA transporter, are present in all thalamic nuclei. The most densely innervated ones are the midline nuclei, intralaminar nuclei (paracentral and parafascicular), and the medial sector of the mediodorsal nucleus (MDm). The ventral motor nuclei and most somatosensory relay nuclei receive a moderate NA innervation. The pulvinar complex receives a heterogeneous innervation. The lateral geniculate nucleus (GL) has the lowest NA innervation. Alpha adrenoceptors were analyzed by in vitro quantitative autoradiography. Alpha-1 receptor densities are higher than Alpha-2 densities. Overall, axonal densities and Alpha adrenoceptor densities coincide; although some mismatches were identified. The nuclei with the highest Alpha-1 values are MDm, the parvocellular part of the ventral posterior medial nucleus, medial pulvinar, and midline nuclei. The nucleus with the lowest Alpha-1 receptor density is GL. Alpha-2 receptor densities are highest in the lateral dorsal, centromedian, medial and inferior pulvinar, and midline nuclei. These results suggest a role for NA in modulating thalamic involvement in consciousness, limbic, cognitive, and executive functions.
Collapse
Affiliation(s)
- Isabel Pérez-Santos
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Calle Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Nicola Palomero-Gallagher
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, 52425 Jülich, Germany.,Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany.,C. & O. Vogt Institute for Brain Research, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Karl Zilles
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, 52425 Jülich, Germany.,C. & O. Vogt Institute for Brain Research, Heinrich-Heine-University, 40225 Düsseldorf, Germany.,JARA-BRAIN, Jülich-Aachen Research Alliance, 52425 Jülich, Germany
| | - Carmen Cavada
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Calle Arzobispo Morcillo 4, 28029 Madrid, Spain
| |
Collapse
|
21
|
Perez DM. Current Developments on the Role of α 1-Adrenergic Receptors in Cognition, Cardioprotection, and Metabolism. Front Cell Dev Biol 2021; 9:652152. [PMID: 34113612 PMCID: PMC8185284 DOI: 10.3389/fcell.2021.652152] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/29/2021] [Indexed: 12/13/2022] Open
Abstract
The α1-adrenergic receptors (ARs) are G-protein coupled receptors that bind the endogenous catecholamines, norepinephrine, and epinephrine. They play a key role in the regulation of the sympathetic nervous system along with β and α2-AR family members. While all of the adrenergic receptors bind with similar affinity to the catecholamines, they can regulate different physiologies and pathophysiologies in the body because they couple to different G-proteins and signal transduction pathways, commonly in opposition to one another. While α1-AR subtypes (α1A, α1B, α1C) have long been known to be primary regulators of vascular smooth muscle contraction, blood pressure, and cardiac hypertrophy, their role in neurotransmission, improving cognition, protecting the heart during ischemia and failure, and regulating whole body and organ metabolism are not well known and are more recent developments. These advancements have been made possible through the development of transgenic and knockout mouse models and more selective ligands to advance their research. Here, we will review the recent literature to provide new insights into these physiological functions and possible use as a therapeutic target.
Collapse
Affiliation(s)
- Dianne M Perez
- The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH, United States
| |
Collapse
|
22
|
Wolf P, Gavins G, Beck‐Sickinger AG, Seitz O. Strategies for Site-Specific Labeling of Receptor Proteins on the Surfaces of Living Cells by Using Genetically Encoded Peptide Tags. Chembiochem 2021; 22:1717-1732. [PMID: 33428317 PMCID: PMC8248378 DOI: 10.1002/cbic.202000797] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/08/2021] [Indexed: 12/14/2022]
Abstract
Fluorescence microscopy imaging enables receptor proteins to be investigated within their biological context. A key challenge is to site-specifically incorporate reporter moieties into proteins without interfering with biological functions or cellular networks. Small peptide tags offer the opportunity to combine inducible labeling with small tag sizes that avoid receptor perturbation. Herein, we review the current state of live-cell labeling of peptide-tagged cell-surface proteins. Considering their importance as targets in medicinal chemistry, we focus on membrane receptors such as G protein-coupled receptors (GPCRs) and receptor tyrosine kinases (RTKs). We discuss peptide tags that i) are subject to enzyme-mediated modification reactions, ii) guide the complementation of reporter proteins, iii) form coiled-coil complexes, and iv) interact with metal complexes. Given our own contributions in the field, we place emphasis on peptide-templated labeling chemistry.
Collapse
Affiliation(s)
- Philipp Wolf
- Faculty of Life SciencesInstitute of BiochemistryLeipzig UniversityBrüderstrasse 3404103LeipzigGermany
| | - Georgina Gavins
- Faculty of Mathematics and Natural SciencesDepartment of ChemistryHumboldt-Universität zu BerlinBrook-Taylor-Str. 212489BerlinGermany
| | - Annette G. Beck‐Sickinger
- Faculty of Life SciencesInstitute of BiochemistryLeipzig UniversityBrüderstrasse 3404103LeipzigGermany
| | - Oliver Seitz
- Faculty of Mathematics and Natural SciencesDepartment of ChemistryHumboldt-Universität zu BerlinBrook-Taylor-Str. 212489BerlinGermany
| |
Collapse
|
23
|
Chmielarz P, Kuśmierczyk J, Rafa-Zabłocka K, Chorązka K, Kowalska M, Satała G, Nalepa I. Antidepressants Differentially Regulate Intracellular Signaling from α1-Adrenergic Receptor Subtypes In Vitro. Int J Mol Sci 2021; 22:ijms22094817. [PMID: 34062902 PMCID: PMC8124549 DOI: 10.3390/ijms22094817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 11/16/2022] Open
Abstract
Currently utilized antidepressants have limited effectiveness and frequently incur undesired effects. Most antidepressants are thought to act via the inhibition of monoamine reuptake; however, direct binding to monoaminergic receptors has been proposed to contribute to both their clinical effectiveness and their side effects, or lack thereof. Among the target receptors of antidepressants, α1‑adrenergic receptors (ARs) have been implicated in depression etiology, antidepressant action, and side effects. However, differences in the direct effects of antidepressants on signaling from the three subtypes of α1-ARs, namely, α1A-, α1B- and α1D‑ARs, have been little explored. We utilized cell lines overexpressing α1A-, α1B- or α1D-ARs to investigate the effects of the antidepressants imipramine (IMI), desipramine (DMI), mianserin (MIA), reboxetine (REB), citalopram (CIT) and fluoxetine (FLU) on noradrenaline-induced second messenger generation by those receptors. We found similar orders of inhibition at α1A-AR (IMI < DMI < CIT < MIA < REB) and α1D‑AR (IMI = DMI < CIT < MIA), while the α1B-AR subtype was the least engaged subtype and was inhibited with low potency by three drugs (MIA < IMI = DMI). In contrast to their direct antagonistic effects, prolonged incubation with IMI and DMI increased the maximal response of the α1B-AR subtype, and the CIT of both the α1A- and the α1B-ARs. Our data demonstrate a complex, subtype-specific modulation of α1-ARs by antidepressants of different groups.
Collapse
Affiliation(s)
- Piotr Chmielarz
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland; (P.C.); (J.K.); (K.R.-Z.); (K.C.); (M.K.)
| | - Justyna Kuśmierczyk
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland; (P.C.); (J.K.); (K.R.-Z.); (K.C.); (M.K.)
| | - Katarzyna Rafa-Zabłocka
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland; (P.C.); (J.K.); (K.R.-Z.); (K.C.); (M.K.)
| | - Katarzyna Chorązka
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland; (P.C.); (J.K.); (K.R.-Z.); (K.C.); (M.K.)
| | - Marta Kowalska
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland; (P.C.); (J.K.); (K.R.-Z.); (K.C.); (M.K.)
| | - Grzegorz Satała
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland;
| | - Irena Nalepa
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland; (P.C.); (J.K.); (K.R.-Z.); (K.C.); (M.K.)
- Correspondence: ; Tel.: +48-12-6623225
| |
Collapse
|
24
|
Andersen DB, Grunddal KV, Pedersen J, Kuhre RE, Lund ML, Holst JJ, Ørskov C. Using a Reporter Mouse to Map Known and Novel Sites of GLP-1 Receptor Expression in Peripheral Tissues of Male Mice. Endocrinology 2021; 162:6122689. [PMID: 33508122 DOI: 10.1210/endocr/bqaa246] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Indexed: 02/07/2023]
Abstract
Glucagon-like peptide-1 receptor (GLP-1R) activation is used in the treatment of diabetes and obesity; however, GLP-1 induces many other physiological effects with unclear mechanisms of action. To identify the cellular targets of GLP-1 and GLP-1 analogues, we generated a Glp1r.tdTomato reporter mouse expressing the reporter protein, tdTomato, in Glp1r-expressing cells. The reporter signal is expressed in all cells where GLP-1R promoter was ever active. To complement this, we histologically mapped tdTomato-fluorescence, and performed Glp-1r mRNA in situ hybridization and GLP-1R immunohistochemistry on the same tissues. In male mice, we found tdTomato signal in mucus neck, chief, and parietal cells of the stomach; Brunner's glands; small intestinal enteroendocrine cells and intraepithelial lymphocytes; and myenteric plexus nerve fibers throughout the gastrointestinal tract. Pancreatic acinar-, β-, and δ cells, but rarely α cells, were tdTomato-positive, as were renal arteriolar smooth muscle cells; endothelial cells of the liver, portal vein, and endocardium; aortal tunica media; and lung type 1 and type 2 pneumocytes. Some thyroid follicular and parafollicular cells displayed tdTomato expression, as did tracheal cartilage chondrocytes, skin fibroblasts, and sublingual gland mucus cells. In conclusion, our reporter mouse is a powerful tool for mapping known and novel sites of GLP-1R expression in the mouse, thus enhancing our understanding of the many target cells and effects of GLP-1 and GLP-1R agonists.
Collapse
Affiliation(s)
- Daniel B Andersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Panum Institute, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kaare V Grunddal
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Panum Institute, Copenhagen, Denmark
| | - Jens Pedersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Panum Institute, Copenhagen, Denmark
- Department of Endocrinology and Nephrology, Nordsjællands Hospital Hillerød, University of Copenhagen, Hillerød, Denmark
| | - Rune E Kuhre
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Panum Institute, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mari L Lund
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Panum Institute, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Cathrine Ørskov
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Panum Institute, Copenhagen, Denmark
| |
Collapse
|
25
|
Fujita S, Yoshida S, Matsuki T, Jaiswal MK, Seki K. The α1-adrenergic receptors in the amygdala regulate the induction of learned despair through protein kinase C-beta signaling. Behav Pharmacol 2021; 32:73-85. [PMID: 33164996 DOI: 10.1097/fbp.0000000000000605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Hyperactivity of amygdala is observed in patients with major depressive disorder. Although the role of α1-adrenoceptor in amygdala on fear memory has been well studied, the role of α1-adrenoceptor in amygdala on depression-like behaviors remains unclear. Therefore, we investigated the effect of α1A-adrenoreceptor in amygdala on despair behavior, evaluated by the immobility time during tail suspension test (TST), pharmacological intervention, and immunohistological methods. C57BL6/J mice given a bilateral intra-amygdala injection of artificial cerebrospinal fluid exhibited an increased duration of immobility in the latter half of both trials of TST with a 24-h interval, a phenomenon known as learned despair. Intra-amygdala injection of WB4101 (1.7 nmol/0.1 µl), an α1 adrenoreceptor antagonist, but not propranolol (250 pmol/0.1 µl), a β-adrenoreceptor antagonist, blocked the induction of learned despair during TST. Immunostaining experiments revealed that ~61-75% of α1A-adrenoreceptor-positive neurons were colocalized with GAD65/67 in amygdala, implying that the α1-adrenoceptors in amygdala may enormously regulate the GABA release. Protein kinase C-beta (PKCβ) was predominantly expressed in the α1A-adrenoreceptor-positive neurons in the BLA, whereas protein kinase C-epsilon (PKCε) was highly expressed with the α1A-adrenoreceptor in the Central nucleus of amygdala. Intra-amygdala injection of ruboxistaurin (10 pmol/0.1 µl), a PKCβ inhibitor, blocked the induction of learned despair during TST, whereas neither TAT-εV1-2 (500 ng/0.1 μl), a cell-permeant PKCε inhibitory peptide, nor HBDDE (50 pmol/0.1 µl), an inhibitor of PKCα and -γ, affected the duration of immobility during TST. These data suggest that the α1-adrenoreceptor in amygdala regulates the induction of learned despair via PKCβ.
Collapse
Affiliation(s)
- Shisui Fujita
- Department of Pharmacology, School of Pharmaceutical Science, Ohu University, Koriyama, Fukushima
| | - Satomi Yoshida
- Department of Pharmacology, School of Pharmaceutical Science, Ohu University, Koriyama, Fukushima
| | - Tohru Matsuki
- Department of Cellular Pathology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai, Aichi, Japan
| | - Manoj Kumar Jaiswal
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kenjiro Seki
- Department of Pharmacology, School of Pharmaceutical Science, Ohu University, Koriyama, Fukushima
| |
Collapse
|
26
|
Lee KS, Navaluna E, Marsh NM, Janezic EM, Hague C. Development of a Novel SNAP-Epitope Tag/Near-Infrared Imaging Assay to Quantify G Protein-Coupled Receptor Degradation in Human Cells. SLAS DISCOVERY 2021; 26:570-578. [PMID: 33402011 DOI: 10.1177/2472555220979793] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
We have developed a novel reporter assay that leverages SNAP-epitope tag/near-infrared (NIR) imaging technology to monitor G protein-coupled receptor (GPCR) degradation in human cell lines. N-terminal SNAP-tagged GPCRs were subcloned and expressed in human embryonic kidney (HEK) 293 cells and then subjected to 24 h of cycloheximide (CHX)-chase degradation assays to quantify receptor degradation half-lives (t1/2) using LICOR NIR imaging-polyacrylamide gel electrophoresis (PAGE) analysis. Thus far, we have used this method to quantify t1/2 for all nine adrenergic (ADRA1A, ADRA1B, ADRA1D, ADRA2A, ADRA2B, ADRA2C, ADRB1, ADRB2, ADRB3), five somatostatin (SSTR1, SSTR2, SSTR3, SSTR4, SSTR5), four chemokine (CXCR1, CXCR2, CXCR3, CXCR5), and three 5-HT2 (5HT2A, 5HT2B, 5HT2C) receptor subtypes. SNAP-GPCR-CHX degradation t1/2 values ranged from 0.52 h (ADRA1D) to 5.5 h (SSTR3). On the contrary, both the SNAP-tag alone and SNAP-tagged and endogenous β-actin were resistant to degradation with CHX treatment. Treatment with the proteasome inhibitor bortezomib produced significant but variable increases in SNAP-GPCR protein expression levels, indicating that SNAP-GPCR degradation primarily occurs through the proteasome. Remarkably, endogenous β2-adrenergic receptor/ADRB2 dynamic mass redistribution functional responses to norepinephrine were significantly decreased following CHX treatment, with a time course equivalent to that observed with the SNAP-ADRB2 degradation assay. We subsequently adapted this assay into a 96-well glass-bottom plate format to facilitate high-throughput GPCR degradation screening. t1/2 values quantified for the α1-adrenergic receptor subtypes (ADRA1A, ADRA1B, ADR1D) using the 96-well-plate format correlated with t1/2 values quantified using NIR-PAGE imaging analysis. In summary, this novel assay permits precise quantitative analysis of GPCR degradation in human cells and can be readily adapted to quantify degradation for any membrane protein of interest.
Collapse
Affiliation(s)
- Kyung-Soon Lee
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, USA
| | - Edelmar Navaluna
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, USA
| | - Nicole M Marsh
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, USA
| | - Eric M Janezic
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, USA
| | - Chris Hague
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
27
|
Domingos-Souza G, Martinez D, Sinkler S, Heesch CM, Kline DD. Alpha adrenergic receptor signaling in the hypothalamic paraventricular nucleus is diminished by the chronic intermittent hypoxia model of sleep apnea. Exp Neurol 2021; 335:113517. [PMID: 33132201 PMCID: PMC7750300 DOI: 10.1016/j.expneurol.2020.113517] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 10/13/2020] [Accepted: 10/20/2020] [Indexed: 01/08/2023]
Abstract
Chronic intermittent hypoxia (CIH) is a model for obstructive sleep apnea. The paraventricular nucleus (PVN) of the hypothalamus has been suggested to contribute to CIH-induced exaggerated cardiorespiratory reflexes, sympathoexcitation and hypertension. This may occur, in part, via activation of the dense catecholaminergic projections to the PVN that originate in the brainstem. However, the contribution of norepinephrine (NE) and activation of its alpha-adrenergic receptors (α-ARs) in the PVN after CIH exposure is unknown. We hypothesized CIH would increase the contribution of catecholaminergic input. To test this notion, we determined the expression of α-AR subtypes, catecholamine terminal density, and synaptic properties of PVN parvocellular neurons in response to α-AR activation in male Sprague-Dawley normoxic (Norm) and CIH exposed rats. CIH decreased mRNA for α1d and α2b AR. Dopamine-β-hydroxylase (DβH) terminals in the PVN were similar between groups. NE and the α1-AR agonist phenylephrine (PE) increased sEPSC frequency after Norm but not CIH. Block of α1-ARs with prazosin alone did not alter sEPSCs after either Norm or CIH but did prevent agonist augmentation of sEPSC frequency following normoxia. These responses to NE were mimicked by PE during action potential block suggesting presynaptic terminal alterations in CIH. Altogether, these results demonstrate that α1-AR activation participates in neuronal responses in Norm, but are attenuated after CIH. These results may provide insight into the cardiovascular, respiratory and autonomic nervous systems alterations in obstructive sleep apnea.
Collapse
Affiliation(s)
- Gean Domingos-Souza
- Dept. of Biomedical Sciences and Dalton Cardiovascular Research Center, Univ. of Missouri, Columbia, MO 65211, United States of America
| | - Diana Martinez
- Dept. of Biomedical Sciences and Dalton Cardiovascular Research Center, Univ. of Missouri, Columbia, MO 65211, United States of America
| | - Steven Sinkler
- Dept. of Biomedical Sciences and Dalton Cardiovascular Research Center, Univ. of Missouri, Columbia, MO 65211, United States of America
| | - Cheryl M Heesch
- Dept. of Biomedical Sciences and Dalton Cardiovascular Research Center, Univ. of Missouri, Columbia, MO 65211, United States of America
| | - David D Kline
- Dept. of Biomedical Sciences and Dalton Cardiovascular Research Center, Univ. of Missouri, Columbia, MO 65211, United States of America.
| |
Collapse
|
28
|
New small molecule fluorescent probes for G protein-coupled receptors: valuable tools for drug discovery. Future Med Chem 2020; 13:63-90. [PMID: 33319586 DOI: 10.4155/fmc-2019-0327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are essential signaling proteins and tractable therapeutic targets. To develop new drug candidates, GPCR drug discovery programs require versatile, sensitive pharmacological tools for ligand binding and compound screening. With the availability of new imaging modalities and proximity-based ligand binding technologies, fluorescent ligands offer many advantages and are increasingly being used, yet labeling small molecules remains considerably more challenging relative to peptides. Focusing on recent fluorescent small molecule studies for family A GPCRs, this review addresses some of the key challenges, synthesis approaches and structure-activity relationship considerations, and discusses advantages of using high-resolution GPCR structures to inform conjugation strategies. While no single approach guarantees successful labeling without loss of affinity or selectivity, the choice of fluorophore, linker type and site of attachment have proved to be critical factors that can significantly affect their utility in drug discovery programs, and as discussed, can sometimes lead to very unexpected results.
Collapse
|
29
|
Zhang J, Simpson PC, Jensen BC. Cardiac α1A-adrenergic receptors: emerging protective roles in cardiovascular diseases. Am J Physiol Heart Circ Physiol 2020; 320:H725-H733. [PMID: 33275531 DOI: 10.1152/ajpheart.00621.2020] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
α1-Adrenergic receptors (ARs) are catecholamine-activated G protein-coupled receptors (GPCRs) that are expressed in mouse and human myocardium and vasculature, and play essential roles in the regulation of cardiovascular physiology. Though α1-ARs are less abundant in the heart than β1-ARs, activation of cardiac α1-ARs results in important biologic processes such as hypertrophy, positive inotropy, ischemic preconditioning, and protection from cell death. Data from the Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial (ALLHAT) indicate that nonselectively blocking α1-ARs is associated with a twofold increase in adverse cardiac events, including heart failure and angina, suggesting that α1-AR activation might also be cardioprotective in humans. Mounting evidence implicates the α1A-AR subtype in these adaptive effects, including prevention and reversal of heart failure in animal models by α1A agonists. In this review, we summarize recent advances in our understanding of cardiac α1A-ARs.
Collapse
Affiliation(s)
- Jiandong Zhang
- McAllister Heart Institute, University of North Carolina, School of Medicine, Chapel Hill, North Carolina
| | - Paul C Simpson
- Department of Medicine and Research Service, San Francisco Veterans Affairs Medical Center and Cardiovascular Research Institute, University of California, San Francisco, California
| | - Brian C Jensen
- McAllister Heart Institute, University of North Carolina, School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
30
|
The Role of Adrenoceptors in the Retina. Cells 2020; 9:cells9122594. [PMID: 33287335 PMCID: PMC7761662 DOI: 10.3390/cells9122594] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 01/16/2023] Open
Abstract
The retina is a part of the central nervous system, a thin multilayer with neuronal lamination, responsible for detecting, preprocessing, and sending visual information to the brain. Many retinal diseases are characterized by hemodynamic perturbations and neurodegeneration leading to vision loss and reduced quality of life. Since catecholamines and respective bindings sites have been characterized in the retina, we systematically reviewed the literature with regard to retinal expression, distribution and function of alpha1 (α1)-, alpha2 (α2)-, and beta (β)-adrenoceptors (ARs). Moreover, we discuss the role of the individual adrenoceptors as targets for the treatment of retinal diseases.
Collapse
|
31
|
Kaykı-Mutlu G, Papazisi O, Palmen M, Danser AHJ, Michel MC, Arioglu-Inan E. Cardiac and Vascular α 1-Adrenoceptors in Congestive Heart Failure: A Systematic Review. Cells 2020; 9:E2412. [PMID: 33158106 PMCID: PMC7694190 DOI: 10.3390/cells9112412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
As heart failure (HF) is a devastating health problem worldwide, a better understanding and the development of more effective therapeutic approaches are required. HF is characterized by sympathetic system activation which stimulates α- and β-adrenoceptors (ARs). The exposure of the cardiovascular system to the increased locally released and circulating levels of catecholamines leads to a well-described downregulation and desensitization of β-ARs. However, information on the role of α-AR is limited. We have performed a systematic literature review examining the role of both cardiac and vascular α1-ARs in HF using 5 databases for our search. All three α1-AR subtypes (α1A, α1B and α1D) are expressed in human and animal hearts and blood vessels in a tissue-dependent manner. We summarize the changes observed in HF regarding the density, signaling and responses of α1-ARs. Conflicting findings arise from different studies concerning the influence that HF has on α1-AR expression and function; in contrast to β-ARs there is no consistent evidence for down-regulation or desensitization of cardiac or vascular α1-ARs. Whether α1-ARs are a therapeutic target in HF remains a matter of debate.
Collapse
Affiliation(s)
- Gizem Kaykı-Mutlu
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, 06560 Ankara, Turkey; (G.K.-M.); (E.A.-I.)
| | - Olga Papazisi
- Department of Cardiothoracic Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (O.P.); (M.P.)
| | - Meindert Palmen
- Department of Cardiothoracic Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (O.P.); (M.P.)
| | - A. H. Jan Danser
- Department of Internal Medicine, Division of Pharmacology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands;
| | - Martin C. Michel
- Department of Pharmacology, Johannes Gutenberg University, 55131 Mainz, Germany
| | - Ebru Arioglu-Inan
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, 06560 Ankara, Turkey; (G.K.-M.); (E.A.-I.)
| |
Collapse
|
32
|
Perez DM. α 1-Adrenergic Receptors in Neurotransmission, Synaptic Plasticity, and Cognition. Front Pharmacol 2020; 11:581098. [PMID: 33117176 PMCID: PMC7553051 DOI: 10.3389/fphar.2020.581098] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/11/2020] [Indexed: 12/14/2022] Open
Abstract
α1-adrenergic receptors are G-Protein Coupled Receptors that are involved in neurotransmission and regulate the sympathetic nervous system through binding and activating the neurotransmitter, norepinephrine, and the neurohormone, epinephrine. There are three α1-adrenergic receptor subtypes (α1A, α1B, α1D) that are known to play various roles in neurotransmission and cognition. They are related to two other adrenergic receptor families that also bind norepinephrine and epinephrine, the β- and α2-, each with three subtypes (β1, β2, β3, α2A, α2B, α2C). Previous studies assessing the roles of α1-adrenergic receptors in neurotransmission and cognition have been inconsistent. This was due to the use of poorly-selective ligands and many of these studies were published before the characterization of the cloned receptor subtypes and the subsequent development of animal models. With the availability of more-selective ligands and the development of animal models, a clearer picture of their role in cognition and neurotransmission can be assessed. In this review, we highlight the significant role that the α1-adrenergic receptor plays in regulating synaptic efficacy, both short and long-term synaptic plasticity, and its regulation of different types of memory. We will also present evidence that the α1-adrenergic receptors, and particularly the α1A-adrenergic receptor subtype, are a potentially good target to treat a wide variety of neurological conditions with diminished cognition.
Collapse
Affiliation(s)
- Dianne M Perez
- The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH, United States
| |
Collapse
|
33
|
Tao YX. Molecular chaperones and G protein-coupled receptor maturation and pharmacology. Mol Cell Endocrinol 2020; 511:110862. [PMID: 32389798 DOI: 10.1016/j.mce.2020.110862] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/15/2022]
Abstract
G protein-coupled receptors (GPCRs) are highly conserved versatile signaling molecules located at the plasma membrane that respond to diverse extracellular signals. They regulate almost all physiological processes in the vertebrates. About 35% of current drugs target these receptors. Mutations in these genes have been identified as causes of numerous diseases. The seven transmembrane domain structure of GPCRs implies that the folding of these transmembrane proteins is extremely complicated and difficult. Indeed, many wild type GPCRs are not folded optimally. The most common defect in genetic diseases caused by GPCR mutations is misfolding and failure to reach the plasma membrane where it functions. General molecular chaperones aid the folding of all proteins, including GPCRs, by preventing aggregation, promoting folding and disaggregating small aggregates. Some GPCRs need additional receptor-specific chaperones to assist their folding. Many of these receptor-specific chaperones interact with additional receptors and alter receptor pharmacology, expanding the understanding of these chaperone proteins.
Collapse
Affiliation(s)
- Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, 36849-5519, USA.
| |
Collapse
|
34
|
Janezic EM, Lauer SML, Williams RG, Chungyoun M, Lee KS, Navaluna E, Lau HT, Ong SE, Hague C. N-glycosylation of α 1D-adrenergic receptor N-terminal domain is required for correct trafficking, function, and biogenesis. Sci Rep 2020; 10:7209. [PMID: 32350295 PMCID: PMC7190626 DOI: 10.1038/s41598-020-64102-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/09/2020] [Indexed: 01/21/2023] Open
Abstract
G protein-coupled receptor (GPCR) biogenesis, trafficking, and function are regulated by post-translational modifications, including N-glycosylation of asparagine residues. α1D-adrenergic receptors (α1D-ARs) - key regulators of central and autonomic nervous system function - contain two putative N-glycosylation sites within the large N-terminal domain at N65 and N82. However, determining the glycosylation state of this receptor has proven challenging. Towards understanding the role of these putative glycosylation sites, site-directed mutagenesis and lectin affinity purification identified N65 and N82 as bona fide acceptors for N-glycans. Surprisingly, we also report that simultaneously mutating N65 and N82 causes early termination of α1D-AR between transmembrane domain 2 and 3. Label-free dynamic mass redistribution and cell surface trafficking assays revealed that single and double glycosylation deficient mutants display limited function with impaired plasma membrane expression. Confocal microscopy imaging analysis and SNAP-tag sucrose density fractionation assays revealed the dual glycosylation mutant α1D-AR is widely distributed throughout the cytosol and nucleus. Based on these novel findings, we propose α1D-AR transmembrane domain 2 acts as an ER localization signal during active protein biogenesis, and that α1D-AR N-terminal glycosylation is required for complete translation of nascent, functional receptor.
Collapse
Affiliation(s)
- Eric M Janezic
- Department of Pharmacology, School of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98185, USA
| | - Sophia My-Linh Lauer
- Department of Pharmacology, School of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98185, USA
| | - Robert George Williams
- Department of Pharmacology, School of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98185, USA
| | - Michael Chungyoun
- Department of Pharmacology, School of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98185, USA
| | - Kyung-Soon Lee
- Department of Pharmacology, School of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98185, USA
| | - Edelmar Navaluna
- Department of Pharmacology, School of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98185, USA
| | - Ho-Tak Lau
- Department of Pharmacology, School of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98185, USA
| | - Shao-En Ong
- Department of Pharmacology, School of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98185, USA
| | - Chris Hague
- Department of Pharmacology, School of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98185, USA.
| |
Collapse
|
35
|
Retinoid acid induced 16 deficiency aggravates colitis and colitis-associated tumorigenesis in mice. Cell Death Dis 2019; 10:958. [PMID: 31862898 PMCID: PMC6925230 DOI: 10.1038/s41419-019-2186-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/02/2019] [Accepted: 12/03/2019] [Indexed: 02/07/2023]
Abstract
Inflammatory bowel disease (IBD) and colitis-associated colorectal cancer (CAC) is a serious health issue, but etiopathological factors remain unclear. Although some studies reported the roles of Retinoid acid induced 16 (RAI16) in the tumorigenesis of hepatocellular carcinoma and PKA signaling, the roles of RAI16 in IBD and CRC are undressed. RAI16−/− mice were generated and the roles of RAI16 were addressed in dextran sodium sulfate (DSS) or azoxymethane (AOM)-DSS induced IBD or CAC mouse models, respectively. At first, RAI16−/− mice were viable, fertile with no apparent defects. Then, it was found that RAI16−/− mice were more susceptibility to colitis induced by DSS than wild type (WT) littermates, which was evaluated by disease activity index and histological score. Furthermore, the expressions of tissues repair associated molecules Cox2, Ereg and MMP-10 were significantly decreased in RAI16−/− colon under DSS treatment. Gut barrier related genes including antimicrobial peptides Reg3b and Reg3g and intestinal mucus genes Muc4, Muc6 and Muc20 were reduced in RAI16−/− colon. These findings indicated that RAI16 may function to affect genes involved in intestinal barrier function and immunoprotective inflammation. Accordingly, RAI16−/− mice displayed significantly increased tumor burden compared with WT mice assessed in CAC model induced by AOM/DSS. Much more Ki67 + nuclei were observed in RAI16−/− tumors suggesting RAI16 to be critical in colonic cell proliferation during tumorigenesis. Conclusively, we demonstrate the roles of RAI16 in colonic inflammation and inflammation-associated tumorigenesis by using a novel RAI16−/− mouse model for the first time.
Collapse
|
36
|
Nascimento-Viana JB, Alcántara-Hernández R, Oliveira-Barros E, Castello Branco LA, Feijó PR, Soares Romeiro LA, Nasciutti LE, Noël F, García-Sáinz JA, Silva CLM. The α1-adrenoceptor-mediated human hyperplastic prostate cells proliferation is impaired by EGF receptor inhibition. Life Sci 2019; 239:117048. [PMID: 31730867 DOI: 10.1016/j.lfs.2019.117048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 10/24/2019] [Accepted: 11/05/2019] [Indexed: 01/12/2023]
Abstract
Benign prostatic hyperplasia (BPH) is an aging-related and progressive disease linked to an up-regulation of α1-adrenoceptors. The participation of EGF receptors (EGFR) in the GPCRs' signalosome has been described but so far data about the contribution of these receptors to prostatic stromal hyperplasia are scanty. We isolated and cultured vimentin-positive prostate stromal cells obtained from BPH patients. According to intracellular Ca2+ measurements, cell proliferation and Western blotting assays, these cultured hyperplastic stromal cells express functional α1-adrenoceptors and EGFR, and proliferate in response to the α1-adrenoceptor agonist phenylephrine. Interestingly, in these cells the inhibition of EGFR signaling with GM6001, CRM197, AG1478 or PD98059 was associated with full blockage of α1-adrenoceptor-mediated cell proliferation, while cell treatment with each inhibitor alone did not alter basal cell growth. Moreover, the co-incubation of AG1478 (EGFR inhibitor) with α1A/α1D-adrenoceptor antagonists showed no additive inhibitory effect. These findings highlight a putative role of EGFR signaling to α1-adrenoceptor-mediated human prostate hyperplasia, suggesting that the inhibition of this transactivation cascade could be useful to reduce BPH progression.
Collapse
Affiliation(s)
| | | | - Eliane Oliveira-Barros
- Cell Biology and Development Research Program, Universidade Federal do Rio de Janeiro, Brazil
| | - Luiza A Castello Branco
- Cell Biology and Development Research Program, Universidade Federal do Rio de Janeiro, Brazil
| | - Priscilla R Feijó
- Laboratory of Biochemical and Molecular Pharmacology, Universidade Federal do Rio de Janeiro, Brazil
| | | | - Luiz Eurico Nasciutti
- Cell Biology and Development Research Program, Universidade Federal do Rio de Janeiro, Brazil
| | - François Noël
- Laboratory of Biochemical and Molecular Pharmacology, Universidade Federal do Rio de Janeiro, Brazil
| | | | | |
Collapse
|
37
|
Alpha1-adrenergic receptor blockade in the ventral tegmental area modulates conditional stimulus-induced cocaine seeking. Neuropharmacology 2019; 158:107680. [DOI: 10.1016/j.neuropharm.2019.107680] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 05/31/2019] [Accepted: 06/20/2019] [Indexed: 11/24/2022]
|
38
|
Hamlett ED, Ledreux A, Gilmore A, Vazey EM, Aston-Jones G, Boger HA, Paredes D, Granholm ACE. Inhibitory designer receptors aggravate memory loss in a mouse model of down syndrome. Neurobiol Dis 2019; 134:104616. [PMID: 31678403 DOI: 10.1016/j.nbd.2019.104616] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 08/06/2019] [Accepted: 09/17/2019] [Indexed: 12/20/2022] Open
Abstract
The pontine nucleus locus coeruleus (LC) is the primary source of noradrenergic (NE) projections to the brain and is important for working memory, attention, and cognitive flexibility. Individuals with Down syndrome (DS) develop Alzheimer's disease (AD) with high penetrance and often exhibit working memory deficits coupled with degeneration of LC-NE neurons early in the progression of AD pathology. Designer receptors exclusively activated by designer drugs (DREADDs) are chemogenetic tools that allow targeted manipulation of discrete neuronal populations in the brain without the confounds of off-target effects. We utilized male Ts65Dn mice (a mouse model for DS), and male normosomic (NS) controls to examine the effects of inhibitory DREADDs delivered via an AAV vector under translational control of the synthetic PRSx8, dopamine β hydroxylase (DβH) promoter. This chemogenetic tool allowed LC inhibition upon administration of the inert DREADD ligand, clozapine-N-oxide (CNO). DREADD-mediated LC inhibition impaired performance in a novel object recognition task and reversal learning in a spatial task. DREADD-mediated LC inhibition gave rise to an elevation of α-adrenoreceptors both in NS and in Ts65Dn mice. Further, microglial markers showed that the inhibitory DREADD stimulation led to increased microglial activation in the hippocampus in Ts65Dn but not in NS mice. These findings strongly suggest that LC signaling is important for intact memory and learning in Ts65Dn mice and disruption of these neurons leads to increased inflammation and dysregulation of adrenergic receptors.
Collapse
Affiliation(s)
- Eric D Hamlett
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA.
| | - Aurélie Ledreux
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO 80208, USA
| | - Anah Gilmore
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO 80208, USA
| | - Elena M Vazey
- Department of Biology, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Gary Aston-Jones
- Rutgers Brain Health Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Heather A Boger
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Daniel Paredes
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO 80208, USA
| | | |
Collapse
|
39
|
Effects of isocorynoxeine, from Uncaria, on lower urinary tract dysfunction caused by benign prostatic hyperplasia via antagonism of α1A-adrenoceptors. Toxicol Appl Pharmacol 2019; 376:95-106. [DOI: 10.1016/j.taap.2019.05.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/21/2019] [Accepted: 05/24/2019] [Indexed: 11/20/2022]
|
40
|
Gudbergsson JM, Duroux M. An evaluation of different Cripto-1 antibodies and their variable results. J Cell Biochem 2019; 121:545-556. [PMID: 31310365 DOI: 10.1002/jcb.29293] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 06/27/2019] [Indexed: 12/14/2022]
Abstract
Cripto-1 is a protein expressed during embryonal development and has been linked to several malignant processes in cancer. Since the discovery of cripto-1 in the late 1980s, it has become a subject of biomarker investigation in several types of cancer which in many cases relies on immunolocalization of cripto-1 using antibodies. Investigating cripto-1 expression and localization in primary glioblastoma cells, we discovered nonspecific binding of cripto-1 antibody to the extracellular matrix Geltrex. A panel of four cripto-1 antibodies was investigated with respect to their binding to the Geltrex matrix and to the cripto-1 positive control cells NTERA2. The cripto-1 expression was varied for the different antibodies with respect to cellular localization and fixation methods. To further elaborate on these findings, we present a systematic review of cripto-1 antibodies found in the literature and highlight some possible cross reactants with data on sequence alignments and structural comparison of EGF domains.
Collapse
Affiliation(s)
- Johann Mar Gudbergsson
- Laboratory of Immunology and Cancer Biology, Institute of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Meg Duroux
- Laboratory of Immunology and Cancer Biology, Institute of Health Science and Technology, Aalborg University, Aalborg, Denmark
| |
Collapse
|
41
|
Use of syngeneic cells expressing membrane-bound GM-CSF as an adjuvant to induce antibodies against native multi-pass transmembrane protein. Sci Rep 2019; 9:9931. [PMID: 31289297 PMCID: PMC6616555 DOI: 10.1038/s41598-019-45160-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/12/2019] [Indexed: 12/23/2022] Open
Abstract
Membrane antigens (mAgs) are important targets for the development of antibody (Ab) drugs. However, native mAgs are not easily prepared, causing difficulties in acquiring functional Abs. In this study, we present a platform in which human mAgs were expressed in native form on cell adjuvants made with membrane-bound cytokines that were then used immunize syngeneic mice directly. The membrane-bound cytokines were used as immune stimulators to enhance specific Ab responses against the desired mAgs. Then, mAgs-expressing xenogeneic cells were used for Ab characterization to reduce non-specific binding. We established cell adjuvants by expressing membrane-bound cytokines (mIL-2, mIL-18, or mGM-CSF) on BALB/3T3 cells, which were effective in stimulating splenocyte proliferation in vitro. We then transiently expressed ecotropic viral integration site 2B (EVI2B) on the adjuvants and used them to directly immunize BALB/c mice. We found that 3T3/mGM-CSF cells stimulated higher specific anti-EVI2B Ab response in the immunized mice than the other cell adjuvants. A G-protein coupled receptor (GPCR), CXCR2, was then transiently expressed on 3T3/mGM-CSF cell adjuvant to immunize mice. The immune serum exhibited relatively higher binding to xenogeneic 293 A/CXCR2 cells than 293 A cells (~3.5-fold). Several hybridoma clones also exhibited selective binding to 293 A/CXCR2 cells. Therefore, the cell adjuvant could preserve the native conformation of mAgs and exhibit anti-mAg Ab stimulatory ability, providing a more convenient and effective method to generate functional Abs, thus possibly accelerating Ab drug development.
Collapse
|
42
|
The monoclonal antibody EPR1614Y against the stem cell biomarker keratin K15 lacks specificity and reacts with other keratins. Sci Rep 2019; 9:1943. [PMID: 30760780 PMCID: PMC6374370 DOI: 10.1038/s41598-018-38163-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 12/20/2018] [Indexed: 12/17/2022] Open
Abstract
Keratin 15 (K15), a type I keratin, which pairs with K5 in epidermis, has been used extensively as a biomarker for stem cells. Two commercial antibodies, LHK15, a mouse monoclonal and EPR1614Y, a rabbit monoclonal, have been widely employed to study K15 expression. Here we report differential reactivity of these antibodies on epithelial cells and tissue sections. Although the two antibodies specifically recognised K15 on western blot, they reacted differently on skin sections and cell lines. LHK15 reacted in patches, whereas EPR1614Y reacted homogenously with the basal keratinocytes in skin sections. In cultured cells, LHK15 did not react with K15 deficient NEB-1, KEB-11, MCF-7 and SW13 cells expressing only exogenous K8 and K18 but reacted when these cells were transduced with K15. On the other hand, EPR1614Y reacted with these cells even though they were devoid of K15. Taken together these results suggest that EPR1614Y recognises a conformational epitope on keratin filaments which can be reconstituted by other keratins as well as by K15. In conclusion, this report highlights that all commercially available antibodies may not be equally specific in identifying the K15 positive stem cell.
Collapse
|
43
|
Bird MF, Guerrini R, Willets JM, Thompson JP, Caló G, Lambert DG. Nociceptin/Orphanin FQ (N/OFQ) conjugated to ATTO594: a novel fluorescent probe for the N/OFQ (NOP) receptor. Br J Pharmacol 2018; 175:4496-4506. [PMID: 30276802 PMCID: PMC6255954 DOI: 10.1111/bph.14504] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/31/2018] [Accepted: 09/04/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE The nociceptin/orphanin FQ (N/OFQ) receptor (NOP) is a member of the opioid receptor family and is involved in a number of physiological responses, pain and immune regulation as examples. In this study, we conjugated a red fluorophore-ATTO594 to the peptide ligand N/OFQ (N/OFQATTO594 ) for the NOP receptor and explored NOP receptor function at high (in recombinant systems) and low (on immune cells) expression. EXPERIMENTAL APPROACH We assessed N/OFQATTO594 receptor binding, selectivity and functional activity in recombinant (CHO) cell lines. Live cell N/OFQATTO594 binding was measured in (i) HEK cells expressing NOP and NOPGFP receptors, (ii) CHO cells expressing the hNOPGαqi5 chimera (to force coupling to measurable Ca2+ responses) and (iii) freshly isolated human polymorphonuclear cells (PMN). KEY RESULTS N/OFQATTO594 bound to NOP receptor with nM affinity and high selectivity. N/OFQATTO594 activated NOP receptor by reducing cAMP formation and increasing Ca2+ levels in CHOhNOPGαqi5 cells. N/OFQATTO594 was also able to visualize NOP receptors at low expression levels on PMN cells. In NOP-GFP-tagged receptors, N/OFQATTO594 was used in a FRET protocol where GFP emission activated ATTO, visualizing ligand-receptor interaction. When the NOPGFP receptor is activated by N/OFQATTO594 , movement of ligand and receptor from the cell surface to the cytosol can be measured. CONCLUSIONS AND IMPLICATIONS In the absence of validated NOP receptor antibodies and issues surrounding the use of radiolabels (especially in low expression systems), these data indicate the utility of N/OFQATTO594 to study a wide range of N/OFQ-driven cellular responses.
Collapse
Affiliation(s)
- M F Bird
- Department of Cardiovascular Sciences, Anaesthesia, Critical Care and Pain Management, Leicester Royal InfirmaryUniversity of LeicesterLeicesterUK
| | - R Guerrini
- Department of Chemical and Pharmaceutical Sciences and LTTAUniversity of FerraraFerraraItaly
| | - J M Willets
- Department of Molecular and Cell BiologyUniversity of LeicesterLeicesterUK
| | - J P Thompson
- Department of Cardiovascular Sciences, Anaesthesia, Critical Care and Pain Management, Leicester Royal InfirmaryUniversity of LeicesterLeicesterUK
| | - G Caló
- Department of Medical Sciences, Section of Pharmacology and National Institute of NeuroscienceUniversity of FerraraFerraraItaly
| | - D G Lambert
- Department of Cardiovascular Sciences, Anaesthesia, Critical Care and Pain Management, Leicester Royal InfirmaryUniversity of LeicesterLeicesterUK
| |
Collapse
|
44
|
|
45
|
Generating a recombinant phosphothreonine-binding domain for a phosphopeptide of the human transcription factor, c-Myc. N Biotechnol 2018; 45:36-44. [PMID: 29763736 DOI: 10.1016/j.nbt.2018.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 04/28/2018] [Accepted: 05/02/2018] [Indexed: 11/21/2022]
Abstract
Transcription factor c-Myc is an oncoprotein that is regulated at the post-translational level through phosphorylation of two conserved residues, Serine 62 (Ser62) and Threonine 58 (Thr58). A highly specific tool capable of recognizing Myc via pThr58 is needed to monitor activation and localization. Through phage display, we have isolated 10 engineered Forkhead-associated (FHA) domains that selectively bind to a phosphothreonine (pThr)-containing peptide (53-FELLPpTPPLSPS-64) segment of human c-Myc. One domain variant was observed to bind to the Myc-pThr58 peptide with a KD value of 800 nM and had >1000-fold discrimination between the phosphorylated and non-phosphorylated peptide. The crystal structure of the engineered FHA Myc-pThr-binding domain (Myc-pTBD) was solved in complex with its cognate ligand. The Myc-pTBD was observed to be structurally similar to the yeast Rad9 FHA1 domain, except that its β4-β5 and β10-β11 loops form a hydrophobic pocket to facilitate the interaction between the domain and the peptide ligand. The Myc-pTBD's specificity for its cognate ligand was demonstrated to be on a par with 3 commercial polyclonal antibodies, suggesting that this recombinant reagent is a viable alternative to antibodies for monitoring Myc regulation.
Collapse
|
46
|
Musayeva A, Manicam C, Steege A, Brochhausen C, Straub BK, Bell K, Pfeiffer N, Gericke A. Role of α 1-adrenoceptor subtypes on corneal epithelial thickness and cell proliferation in mice. Am J Physiol Cell Physiol 2018; 315:C757-C765. [PMID: 30257104 DOI: 10.1152/ajpcell.00314.2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adrenergic stimuli are important for corneal epithelial structure and healing. The purpose of the present study was to examine the hypothesis that the lack of a single α1-adrenoceptor (α1-AR) subtype affects corneal epithelial thickness and cell proliferation. Expression levels of α1-AR mRNA were determined in mouse cornea using real-time PCR. In mice devoid of one of the three α1-AR subtypes (α1A-AR-/-, α1B-AR-/-, α1D-AR-/-) and in wild-type controls, thickness of individual corneal layers, the number of epithelial cell layers, and average epithelial cell size were determined in cryosections. Endothelial cell density and morphology were calculated in corneal explants, and epithelial cell proliferation rate was determined with immunofluorescence microscopy. Moreover, the ultrastructure of the corneal epithelium was examined by transmission electron microscopy. Messenger RNA for all three α1-AR subtypes was expressed in whole cornea and in corneal epithelium from wild-type mice with a rank order of abundance of α1A ≥ α1B > α1D. In contrast, no α1-AR mRNA was detected in the stroma, and only α1B-AR mRNA was found in the Descemet endothelial complex. Remarkably, corneal epithelial thickness and mean epithelial cell size were reduced in α1A-AR-/- mice. Our findings suggest that the α1A-AR exerts growth effects in mouse corneal epithelial cells.
Collapse
Affiliation(s)
- Aytan Musayeva
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz , Mainz , Germany
| | - Caroline Manicam
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz , Mainz , Germany
| | - Andreas Steege
- Department of Internal Medicine II, University Medical Center Regensburg , Regensburg , Germany
| | - Christoph Brochhausen
- Institute of Pathology, University Medical Center, Johannes Gutenberg University Mainz , Mainz , Germany.,Institute of Pathology, University Medical Center Regensburg , Regensburg , Germany
| | - Beate K Straub
- Institute of Pathology, University Medical Center, Johannes Gutenberg University Mainz , Mainz , Germany
| | - Katharina Bell
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz , Mainz , Germany
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz , Mainz , Germany
| | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz , Mainz , Germany
| |
Collapse
|
47
|
Abstract
Validation of antibodies and other protein binders is a subject of pressing concern for the research community and one which is uppermost in the minds of all who use antibodies as research and diagnostic reagents. Assessing an antibody's fitness for purpose includes accurate ascertainment of its target specificity and suitability for the envisaged task. Moreover, standardised procedures are essential to guarantee sample quality in testing procedures. The problem of defining precise standards for antibody validation has engendered much debate in recent publications and meetings, but gradually a consensus is emerging. At the 8th Alpbach Affinity Proteomics workshop (March 2017), a panel of leaders in the antibody field discussed suggestions which could bring this complex but essential issue a step nearer to a resolution. 'Alpbach recommendations' for best practice include tailoring binder validation processes according to the intended applications and promoting greater transparency in publications and in the information available from commercial antibody developers/providers. A single approach will not fit all applications and end users must ensure that the reported validation holds for their specific requirements, highlighting the need for adequate training in the fundamentals of antibody characterisation and validation across the user community.
Collapse
Affiliation(s)
- Michael J Taussig
- Cambridge Protein Arrays Ltd., Babraham Research Campus, Cambridge, CB22 3AT, UK.
| | - Cláudia Fonseca
- Cambridge Protein Arrays Ltd., Babraham Research Campus, Cambridge, CB22 3AT, UK.
| | - James S Trimmer
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, CA, 95616, USA; Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, CA, 95616, USA.
| |
Collapse
|
48
|
Dahl EF, Wu SC, Healy CL, Harsch BA, Shearer GC, O'Connell TD. Subcellular compartmentalization of proximal Gα q-receptor signaling produces unique hypertrophic phenotypes in adult cardiac myocytes. J Biol Chem 2018; 293:8734-8749. [PMID: 29610273 DOI: 10.1074/jbc.ra118.002283] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/13/2018] [Indexed: 12/12/2022] Open
Abstract
G protein-coupled receptors that signal through Gαq (Gq receptors), such as α1-adrenergic receptors (α1-ARs) or angiotensin receptors, share a common proximal signaling pathway that activates phospholipase Cβ1 (PLCβ1), which cleaves phosphatidylinositol 4,5-bisphosphate (PIP2) to produce inositol 1,4,5-trisphosphate (IP3) and diacylglycerol. Despite these common proximal signaling mechanisms, Gq receptors produce distinct physiological responses, yet the mechanistic basis for this remains unclear. In the heart, Gq receptors are thought to induce myocyte hypertrophy through a mechanism termed excitation-transcription coupling, which provides a mechanistic basis for compartmentalization of calcium required for contraction versus IP3-dependent intranuclear calcium required for hypertrophy. Here, we identified subcellular compartmentalization of Gq-receptor signaling as a mechanistic basis for unique Gq receptor-induced hypertrophic phenotypes in cardiac myocytes. We show that α1-ARs co-localize with PLCβ1 and PIP2 at the nuclear membrane. Further, nuclear α1-ARs induced intranuclear PLCβ1 activity, leading to histone deacetylase 5 (HDAC5) export and a robust transcriptional response (i.e. significant up- or down-regulation of 806 genes). Conversely, we found that angiotensin receptors localize to the sarcolemma and induce sarcolemmal PLCβ1 activity, but fail to promote HDAC5 nuclear export, while producing a transcriptional response that is mostly a subset of α1-AR-induced transcription. In summary, these results link Gq-receptor compartmentalization in cardiac myocytes to unique hypertrophic transcription. They suggest a new model of excitation-transcription coupling in adult cardiac myocytes that accounts for differential Gq-receptor localization and better explains distinct physiological functions of Gq receptors.
Collapse
Affiliation(s)
| | - Steven C Wu
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota 55455 and
| | - Chastity L Healy
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota 55455 and
| | - Brian A Harsch
- the Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania 16802
| | - Gregory C Shearer
- the Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania 16802
| | - Timothy D O'Connell
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota 55455 and
| |
Collapse
|
49
|
Lund-Johansen F, Browning MD. Should we ignore western blots when selecting antibodies for other applications? Nat Methods 2018; 14:215. [PMID: 28245212 DOI: 10.1038/nmeth.4192] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Fridtjof Lund-Johansen
- K G Jebsen Center for Cancer Immunotherapy, Oslo University Hospital, Oslo, Norway.,Department of Immunology, Oslo University Hospital, Oslo, Norway
| | | |
Collapse
|
50
|
Affiliation(s)
- Victoria A. Meliopoulos
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- * E-mail:
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| |
Collapse
|