1
|
Liu Q, Tang Z, Qian Y, Wang C, Kong C, Li M, Geng X, Zhang Y, Cheng X, Ren C, Wang K, Bai L, Wang L, Jiang D, Wang S, Liu X, Xia P. Eukaryotic ADCY7 catalyzes the production of c-di-AMP to activate the NLRP3 inflammasome. Nat Chem Biol 2025:10.1038/s41589-025-01919-y. [PMID: 40419769 DOI: 10.1038/s41589-025-01919-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 04/21/2025] [Indexed: 05/28/2025]
Abstract
Toll-like receptor 9 (TLR9) agonists cause activation of nucleotide-binding domain, leucine-rich repeat protein 3 (NLRP3) inflammasomes but the mechanism is not clear. We found that there is a second signal downstream of TLR9 that induces NLRP3 inflammasome activation. Through screening, adenylate cyclase 7 (ADCY7) was found to be an essential regulator of this process. In cells with Adcy7 deficiency, TLR9 agonists were no longer able to activate the NLRP3 inflammasome. ADCY7 not only catalyzes the generation of cyclic adenosine monophosphate (cAMP) but also catalyzes the synthesis of its dimeric form (c-di-AMP). Moreover, c-di-AMP promotes assembly and maturation of the inflammasome by directly binding to NLRP3. Cells with Adcy7 deletion or mutations impacting enzymatic activity cannot produce c-di-AMP. The survival of Adcy7-deficient mice in acute liver injury was also improved. In summary, we found that ADCY7 is required for NLRP3 inflammasome activation downstream of TLR9 by catalyzing the generation of c-di-AMP, which may serve as a target for controlling inflammatory responses in sterile infections.
Collapse
Affiliation(s)
- Qiannv Liu
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
- Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing, China
| | - Zhiheng Tang
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yan Qian
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
- Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing, China
| | - Chunlei Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
- Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing, China
| | - Chun Kong
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
- Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing, China
| | - Mengqian Li
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
- Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing, China
| | - Xiangyang Geng
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
- Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing, China
| | - Yan Zhang
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
- Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing, China
| | - Xiangyun Cheng
- Department of Sports Medicine, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Institute of Sports Medicine of Peking University, Beijing, China
| | - Chao Ren
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Kai Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
- Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing, China
| | - Lin Bai
- Department of Biophysics, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Lin Wang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Dong Jiang
- Department of Sports Medicine, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Institute of Sports Medicine of Peking University, Beijing, China
| | - Shuo Wang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
| | - Xiaoyun Liu
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China.
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University, Beijing, China.
- Department of Infectious Diseases, Peking University Third Hospital, Beijing, China.
| | - Pengyan Xia
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China.
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China.
- Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing, China.
- PUHSC Primary Immunodeficiency Research Center, Peking University, Beijing, China.
| |
Collapse
|
2
|
Szukiewicz D. Potential Therapeutic Exploitation of G Protein-Coupled Receptor 120 (GPR120/FFAR4) Signaling in Obesity-Related Metabolic Disorders. Int J Mol Sci 2025; 26:2501. [PMID: 40141148 PMCID: PMC11941992 DOI: 10.3390/ijms26062501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/08/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
The increasing prevalence of overweight and obesity not only in adults but also among children and adolescents has become one of the most alarming health problems worldwide. Metabolic disorders accompanying fat accumulation during pathological weight gain induce chronic low-grade inflammation, which, in a vicious cycle, increases the immune response through pro-inflammatory changes in the cytokine (adipokine) profile. Obesity decreases life expectancy, largely because obese individuals are at an increased risk of many medical complications, often referred to as metabolic syndrome, which refers to the co-occurrence of insulin resistance (IR), impaired glucose tolerance, type 2 diabetes (T2D), atherogenic dyslipidemia, hypertension, and premature ischemic heart disease. Metabotropic G protein-coupled receptors (GPCRs) constitute the most numerous and diverse group of cell surface transmembrane receptors in eukaryotes. Among the GPCRs, researchers are focusing on the connection of G protein-coupled receptor 120 (GPR120), also known as free fatty acid receptor 4 (FFAR4), with signaling pathways regulating the inflammatory response and insulin sensitivity. This review presents the current state of knowledge concerning the involvement of GPR120 in anti-inflammatory and metabolic signaling. Since both inflammation in adipose tissue and insulin resistance are key problems in obesity, there is a rationale for the development of novel, GPR120-based therapies for overweight and obese individuals. The main problems associated with introducing this type of treatment into clinical practice are also discussed.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
3
|
Erboz A, Kesekler E, Gentili PL, Uversky VN, Coskuner-Weber O. Electromagnetic radiation and biophoton emission in neuronal communication and neurodegenerative diseases. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 195:87-99. [PMID: 39732343 DOI: 10.1016/j.pbiomolbio.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/08/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
The intersection of electromagnetic radiation and neuronal communication, focusing on the potential role of biophoton emission in brain function and neurodegenerative diseases is an emerging research area. Traditionally, it is believed that neurons encode and communicate information via electrochemical impulses, generating electromagnetic fields detectable by EEG and MEG. Recent discoveries indicate that neurons may also emit biophotons, suggesting an additional communication channel alongside the regular synaptic interactions. This dual signaling system is analyzed for its potential in synchronizing neuronal activity and improving information transfer, with implications for brain-like computing systems. The clinical relevance is explored through the lens of neurodegenerative diseases and intrinsically disordered proteins, where oxidative stress may alter biophoton emission, offering clues for pathological conditions, such as Alzheimer's and Parkinson's diseases. The potential therapeutic use of Low-Level Laser Therapy (LLLT) is also examined for its ability to modulate biophoton activity and mitigate oxidative stress, presenting new opportunities for treatment. Here, we invite further exploration into the intricate roles the electromagnetic phenomena play in brain function, potentially leading to breakthroughs in computational neuroscience and medical therapies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Aysin Erboz
- Molecular Biotechnology, Turkish-German University, Sahinkaya Caddesi No. 106, Beykoz, Istanbul, 34820, Turkey
| | - Elif Kesekler
- Molecular Biotechnology, Turkish-German University, Sahinkaya Caddesi No. 106, Beykoz, Istanbul, 34820, Turkey
| | - Pier Luigi Gentili
- Department of Chemistry, Biology, and Biotechnology, Università degli Studi di Perugia, 06123, Perugia, Italy.
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Institute, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd., MDC07, Tampa, FL 33612, USA.
| | - Orkid Coskuner-Weber
- Molecular Biotechnology, Turkish-German University, Sahinkaya Caddesi No. 106, Beykoz, Istanbul, 34820, Turkey.
| |
Collapse
|
4
|
Zhang Y, Mazal H, Mandala VS, Pérez-Mitta G, Sondoghdar V, Haselwandter CA, MacKinnon R. Higher-order transient membrane protein structures. Proc Natl Acad Sci U S A 2025; 122:e2421275121. [PMID: 39739811 PMCID: PMC11725870 DOI: 10.1073/pnas.2421275121] [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: 10/16/2024] [Accepted: 11/20/2024] [Indexed: 01/02/2025] Open
Abstract
This study shows that five membrane proteins-three GPCRs, an ion channel, and an enzyme-form self-clusters under natural expression levels in a cardiac-derived cell line. The cluster size distributions imply that these proteins self-oligomerize reversibly through weak interactions. When the concentration of the proteins is increased through heterologous expression, the cluster size distributions approach a critical distribution at which point a phase transition occurs, yielding larger bulk phase clusters. A thermodynamic model like that explaining micellization of amphiphiles and lipid membrane formation accounts for this behavior. We propose that many membrane proteins exist as oligomers that form through weak interactions, which we call higher-order transient structures (HOTS). The key characteristics of HOTS are transience, molecular specificity, and a monotonically decreasing size distribution that may become critical at high concentrations. Because molecular specificity invokes self-recognition through protein sequence and structure, we propose that HOTS are genetically encoded supramolecular units.
Collapse
Affiliation(s)
- Yuxi Zhang
- Laboratory of Molecular Neurobiology and Biophysics, The Rockefeller University, New York, NY10065
- HHMI, The Rockefeller University, New York, NY10065
| | - Hisham Mazal
- Max Planck Institute for the Science of Light, Erlangen91058, Germany
- Max-Planck-Zentrum für Physik und Medizin, Erlangen91058, Germany
| | - Venkata Shiva Mandala
- Laboratory of Molecular Neurobiology and Biophysics, The Rockefeller University, New York, NY10065
- HHMI, The Rockefeller University, New York, NY10065
| | - Gonzalo Pérez-Mitta
- Laboratory of Molecular Neurobiology and Biophysics, The Rockefeller University, New York, NY10065
- HHMI, The Rockefeller University, New York, NY10065
| | - Vahid Sondoghdar
- Max Planck Institute for the Science of Light, Erlangen91058, Germany
- Max-Planck-Zentrum für Physik und Medizin, Erlangen91058, Germany
- Department of Physics, Friedrich Alexander University Erlangen-Nürnberg, Erlangen91058, Germany
| | - Christoph A. Haselwandter
- Department of Physics and Astronomy, University of Southern California, Los Angeles, CA90089
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA90089
| | - Roderick MacKinnon
- Laboratory of Molecular Neurobiology and Biophysics, The Rockefeller University, New York, NY10065
- HHMI, The Rockefeller University, New York, NY10065
| |
Collapse
|
5
|
Zhang Y, MacKinnon R. Higher-order transient structures and the principle of dynamic connectivity in membrane signaling. Proc Natl Acad Sci U S A 2025; 122:e2421280121. [PMID: 39739805 PMCID: PMC11725812 DOI: 10.1073/pnas.2421280121] [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: 10/15/2024] [Accepted: 11/22/2024] [Indexed: 01/02/2025] Open
Abstract
We examine the role of higher-order transient structures (HOTS) in M2R regulation of GIRK channels. Electron microscopic membrane protein location maps show that both proteins form HOTS that exhibit a statistical bias to be near each other. Theoretical calculations and electrophysiological measurements suggest that channel activity is isolated near larger M2R HOTS. By invoking weak interactions that permit transient binding of M2R to M2R and GIRK to GIRK (i-i interactions) and M2R to GIRK (i-j interactions), the distribution patterns and electrophysiological properties of HL-1 cells are replicated in a reaction-diffusion simulation. We propose the principle of dynamic connectivity to explain communication between protein components of a membrane signaling pathway. Dynamic connectivity is mediated by weak, transient interactions between proteins. HOTS created by weak i-i interactions, and statistical biases created by weak i-j interactions promoted by the multivalence of HOTS, are the key elements of dynamic connectivity.
Collapse
Affiliation(s)
- Yuxi Zhang
- Laboratory of Molecular Neurobiology and Biophysics, The Rockefeller University, New York, NY10065
- HHMI, The Rockefeller University, New York, NY10065
| | - Roderick MacKinnon
- Laboratory of Molecular Neurobiology and Biophysics, The Rockefeller University, New York, NY10065
- HHMI, The Rockefeller University, New York, NY10065
| |
Collapse
|
6
|
Lauritano C, Bazzani E, Montuori E, Bolinesi F, Mangoni O, Riccio G, Buondonno A, Saggiomo M. Salinity Stress Acclimation Strategies in Chlamydomonas sp. Revealed by Physiological, Morphological and Transcriptomic Approaches. Mar Drugs 2024; 22:351. [PMID: 39195467 DOI: 10.3390/md22080351] [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: 06/16/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024] Open
Abstract
Climate changes may include variations in salinity concentrations at sea by changing ocean dynamics. These variations may be especially challenging for marine photosynthetic organisms, affecting their growth and distribution. Chlamydomonas spp. are ubiquitous and are often found in extreme salinity conditions. For this reason, they are considered good model species to study salinity adaptation strategies. In the current study, we used an integrated approach to study the Chlamydomonas sp. CCMP225 response to salinities of 20‱ and 70‱, by combining physiological, morphological, and transcriptomic analyses, and comparing differentially expressed genes in the exponential and stationary growth phases under the two salinity conditions. The results showed that the strain is able to grow under all tested salinity conditions and maintains a surprisingly high photosynthetic efficiency even under high salinities. However, at the highest salinity condition, the cells lose their flagella. The transcriptomic analysis highlighted the up- or down-regulation of specific gene categories, helping to identify key genes responding to salinity stress. Overall, the findings may be of interest to the marine biology, ecology, and biotechnology communities, to better understand species adaptation mechanisms under possible global change scenarios and the potential activation of enzymes involved in the synthesis of bioactive molecules.
Collapse
Affiliation(s)
- Chiara Lauritano
- Ecosustainable Marine Biotechnology Department, Stazione Zoologica Anton Dohrn, Via Acton, 80133 Naples, Italy
| | - Emma Bazzani
- Research Infrastructure for Marine Biological Resources Department, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Naples, Italy
- Smurfit Institute of Genetics, School of Genetics and Microbiology, Trinity College Dublin, College Green, Dublin 2, D02 VF25 Dublin, Ireland
| | - Eleonora Montuori
- Ecosustainable Marine Biotechnology Department, Stazione Zoologica Anton Dohrn, Via Acton, 80133 Naples, Italy
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Francesco Bolinesi
- Department of Biology, University of Naples Federico II, Via Cinthia 21, 80126 Naples, Italy
- CoNISMa, Piazzale Flaminio, 9, 00196 Roma, Italy
| | - Olga Mangoni
- Department of Biology, University of Naples Federico II, Via Cinthia 21, 80126 Naples, Italy
- CoNISMa, Piazzale Flaminio, 9, 00196 Roma, Italy
| | - Gennaro Riccio
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Naples, Italy
| | - Angela Buondonno
- Research Infrastructure for Marine Biological Resources Department, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Naples, Italy
| | - Maria Saggiomo
- Research Infrastructure for Marine Biological Resources Department, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Naples, Italy
| |
Collapse
|
7
|
Sakamoto E, Kitase Y, Fitt AJ, Zhu Z, Awad K, Brotto M, White KE, Welc SS, Bergwitz C, Bonewald LF. Both enantiomers of β-aminoisobutyric acid BAIBA regulate Fgf23 via MRGPRD receptor by activating distinct signaling pathways in osteocytes. Cell Rep 2024; 43:114397. [PMID: 38935499 PMCID: PMC11350516 DOI: 10.1016/j.celrep.2024.114397] [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: 12/13/2023] [Revised: 04/24/2024] [Accepted: 06/07/2024] [Indexed: 06/29/2024] Open
Abstract
With exercise, muscle and bone produce factors with beneficial effects on brain, fat, and other organs. Exercise in mice increased fibroblast growth factor 23 (FGF23), urine phosphate, and the muscle metabolite L-β-aminoisobutyric acid (L-BAIBA), suggesting that L-BAIBA may play a role in phosphate metabolism. Here, we show that L-BAIBA increases in serum with exercise and elevates Fgf23 in osteocytes. The D enantiomer, described to be elevated with exercise in humans, can also induce Fgf23 but through a delayed, indirect process via sclerostin. The two enantiomers both signal through the same receptor, Mas-related G-protein-coupled receptor type D, but activate distinct signaling pathways; L-BAIBA increases Fgf23 through Gαs/cAMP/PKA/CBP/β-catenin and Gαq/PKC/CREB, whereas D-BAIBA increases Fgf23 indirectly through sclerostin via Gαi/NF-κB. In vivo, both enantiomers increased Fgf23 in bone in parallel with elevated urinary phosphate excretion. Thus, exercise-induced increases in BAIBA and FGF23 work together to maintain phosphate homeostasis.
Collapse
Affiliation(s)
- Eijiro Sakamoto
- Department of Anatomy, Cell Biology, & Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Yukiko Kitase
- Department of Anatomy, Cell Biology, & Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN 46202, USA.
| | - Alexander J Fitt
- Department of Anatomy, Cell Biology, & Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Zewu Zhu
- Section of Endocrinology and Metabolism, Yale School of Medicine, New Haven, CT 06519, USA
| | - Kamal Awad
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas-Arlington, Arlington, TX 76019, USA
| | - Marco Brotto
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas-Arlington, Arlington, TX 76019, USA
| | - Kenneth E White
- Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Department of Molecular and Medical Genetics, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Steven S Welc
- Department of Anatomy, Cell Biology, & Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Clemens Bergwitz
- Section of Endocrinology and Metabolism, Yale School of Medicine, New Haven, CT 06519, USA
| | - Lynda F Bonewald
- Department of Anatomy, Cell Biology, & Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Department of Orthopaedic Surgery, School of Medicine, Indiana University, Indianapolis, IN 46202, USA.
| |
Collapse
|
8
|
Dong H, Wang Y, Zhi T, Guo H, Guo Y, Liu L, Yin Y, Shi J, He B, Hu L, Jiang G. Construction of protein-protein interaction network in sulfate-reducing bacteria: Unveiling of global response to Hg. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 351:124048. [PMID: 38714230 DOI: 10.1016/j.envpol.2024.124048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 05/09/2024]
Abstract
Sulfate-reducing bacteria (SRB) play pivotal roles in the biotransformation of mercury (Hg). However, unrevealed global responses of SRB to Hg have restricted our understanding of details of Hg biotransformation processes. The absence of protein-protein interaction (PPI) network under Hg stimuli has been a bottleneck of proteomic analysis for molecular mechanisms of Hg transformation. This study constructed the first comprehensive PPI network of SRB in response to Hg, encompassing 67 connected nodes, 26 independent nodes, and 121 edges, covering 93% of differentially expressed proteins from both previous studies and this study. The network suggested that proteomic changes of SRB in response to Hg occurred globally, including microbial metabolism in diverse environments, carbon metabolism, nucleic acid metabolism and translation, nucleic acid repair, transport systems, nitrogen metabolism, and methyltransferase activity, partial of which could cover the known knowledge. Antibiotic resistance was the original response revealed by this network, providing insights into of Hg biotransformation mechanisms. This study firstly provided the foundational network for a comprehensive understanding of SRB's responses to Hg, convenient for exploration of potential targets for Hg biotransformation. Furthermore, the network indicated that Hg enhances the metabolic activities and modification pathways of SRB to maintain cellular activities, shedding light on the influences of Hg on the carbon, nitrogen, and sulfur cycles at the cellular level.
Collapse
Affiliation(s)
- Hongzhe Dong
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100049, China; Sino-Danish Centre for Education and Research, Beijing, 100049, China
| | - Yuchuan Wang
- Hebei Key Laboratory for Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, 063210, China
| | - Tingting Zhi
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Hua Guo
- School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Yingying Guo
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Lihong Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Yongguang Yin
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Jianbo Shi
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; School of Environment and Health, Jianghan University, Wuhan, 430056, China
| | - Bin He
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Ligang Hu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100049, China; Sino-Danish Centre for Education and Research, Beijing, 100049, China; School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| |
Collapse
|
9
|
Thomas P, Pang Y, Dong J. Ligand-independent signaling and migration of breast cancer cells expressing membrane androgen receptor, ZIP9 (SLC39A9). Mol Cell Endocrinol 2023; 578:112060. [PMID: 37660782 DOI: 10.1016/j.mce.2023.112060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 08/28/2023] [Accepted: 09/01/2023] [Indexed: 09/05/2023]
Abstract
Zinc transporter ZIP9 is also a membrane androgen receptor that mediates androgen-dependent zinc and G-protein signaling to modulate tumorigenic responses in cancer cells. It is unclear whether unliganded ZIP9 causes similar responses. ZIP9 overexpression in MDA-MB-231 breast cancer cells (ZIP9 cells) increased zinc levels and cell migration/invasion which was mimicked with a zinc ionophore and attenuated with a zinc chelator, suggesting these tumorigenic responses are zinc-dependent. Expression of migration markers MYL9 and CYR61 was elevated in ZIP9 cells and further increased together with cell migration by forskolin treatment and blocked with H-89, indicating they are mediated through an AC/PKA pathway. Knockdown of ZIP9 expression in MDA-MB-468 cells decreased cell migration/invasion, migration markers and zinc levels, confirming similar roles of unliganded ZIP9 in another breast cancer cell line. Testosterone treatment further increased migration, biomarker expression and zinc in ZIP9 cells, suggesting it may act through similar pathways to induce tumorigenic responses.
Collapse
Affiliation(s)
- Peter Thomas
- Marine Science Institute, University of Texas at Austin. Port Aransas, Texas, 78373, USA.
| | - Yefei Pang
- Marine Science Institute, University of Texas at Austin. Port Aransas, Texas, 78373, USA
| | - Jing Dong
- Marine Science Institute, University of Texas at Austin. Port Aransas, Texas, 78373, USA
| |
Collapse
|
10
|
Higuchi T, Takagi K, Tochimoto A, Ichimura Y, Hirose H, Sawada T, Shibata N, Harigai M, Kawaguchi Y. Antifibrotic effect of apremilast in systemic sclerosis dermal fibroblasts and bleomycin-induced mouse model. Sci Rep 2023; 13:19378. [PMID: 37938601 PMCID: PMC10632419 DOI: 10.1038/s41598-023-46737-1] [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: 02/21/2023] [Accepted: 11/04/2023] [Indexed: 11/09/2023] Open
Abstract
Phosphodiesterase (PDE) 4 inhibitors have been reported to suppress the progression of dermal fibrosis in patients with systemic sclerosis (SSc); however, the precise mechanisms remain to be elucidated. Therefore, we conducted experiments focusing on the antifibrotic and anti-inflammatory effects of apremilast using dermal fibroblasts derived from patients with SSc and an SSc mouse model. Dermal fibroblasts derived from healthy controls and patients with SSc were incubated with apremilast in the presence or absence of 10 ng/ml transforming growth factor (TGF)-β1 for the measurement of intracellular cAMP levels and evaluation of mRNA and protein expression. A bleomycin-induced dermal fibrosis mouse model was used to evaluate the inhibitory effects of apremilast on the progression of dermal fibrosis. Intracellular cAMP levels were significantly reduced in dermal fibroblasts derived from patients with SSc compared with those derived from healthy controls. Apremilast reduced the mRNA expression of profibrotic markers and the protein expression of type I collagen and Cellular Communication Network Factor 2 (CCN2) in dermal fibroblasts. Additionally, apremilast inhibited the progression of dermal fibrosis in mice, partly by acting on T cells. These results suggest that apremilast may be a potential candidate for treating dermal fibrosis in SSc.
Collapse
Affiliation(s)
- Tomoaki Higuchi
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan.
- Division of Multidisciplinary Management of Rheumatic Diseases, Tokyo Women's Medical University School of Medicine, 8-1, Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan.
| | - Kae Takagi
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Akiko Tochimoto
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Yuki Ichimura
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
- Department of Dermatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hikaru Hirose
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Tatsuo Sawada
- Department of Pathology, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Noriyuki Shibata
- Department of Pathology, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Masayoshi Harigai
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Yasushi Kawaguchi
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| |
Collapse
|
11
|
Xie Z, Fox AP. Rapid emergence from dexmedetomidine sedation in Sprague Dawley rats by repurposing an α 2-adrenergic receptor competitive antagonist in combination with caffeine. BMC Anesthesiol 2023; 23:39. [PMID: 36721095 PMCID: PMC9890710 DOI: 10.1186/s12871-023-01986-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 01/16/2023] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND The α2 adrenergic receptor agonist dexmedetomidine is an important intravenous sedative with analgesic properties. Currently available dexmedetomidine reversal agents, like the α2-receptor antagonist atipamezole, cause serious adverse effects at the large dosages required for effective reversal; they are not used clinically. Without reversal agents, emergence times from dexmedetomidine sedation are slow. In this study we tested the ability of low-dose atipamezole, in combination with caffeine, to reverse dexmedetomidine sedation. The low dose of atipamezole employed should not be associated with unwanted effects. METHODS Two different sedation protocols were employed. In the first protocol, a bolus of dexmedetomidine was rapidly applied and the drug was allowed to equilibrate for 10 min before rats received either saline (as control) or low-dose atipamezole with caffeine. Following this procedure, rats were placed on their backs. Emergence from sedation was the time for rats to recover their righting reflex and stand with 4 paws on the floor. A second sedation protocol simulated a pediatric magnetic resonance imaging (MRI) scan. Adult rats were sedated with dexmedetomidine for one hour followed by 30 min with both dexmedetomidine and propofol. At the end of 90 min, rats received either saline (control) or a combination of low-dose atipamezole, and caffeine. Recovery of the righting reflex was used as a proxy for emergence from sedation. RESULTS Emergence from sedation, the time for rats to recover their righting reflex, decreased by ~ 90% when using an atipamezole dose ~ 20 fold lower than manufacturer's recommendation, supplemented with caffeine. Using an atipamezole dose ~ tenfold lower than recommended, with caffeine, emergence times decreased by ~ 97%. A different stimulant, forskolin, when tested, was as effective as caffeine. For the MRI simulation, emergence times were decreased by ~ 93% by low-dose atipamezole with caffeine. CONCLUSIONS Low dose atipamezole with caffeine was effective at reversing dexmedetomidine sedation. Emergence was rapid and the rats regained not only their righting reflex but also their balance and their ability to carry out complex behaviors. These findings suggest that the combination of low dose atipamezole with caffeine may permit rapid clinical reversal of dexmedetomidine without unwanted effects.
Collapse
Affiliation(s)
- Zheng Xie
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, IL, USA
| | - Aaron P Fox
- Department of Neurobiology, Pharmacology and Physiology, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
12
|
Hou J, He M, Chen Q, Liang S. LncRNA H19 acts as miR-301a-3p sponge to alleviate lung injury in mice with sepsis by regulating Adcy1. Immunopharmacol Immunotoxicol 2022; 44:565-573. [PMID: 35438054 DOI: 10.1080/08923973.2022.2067045] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND The abnormal expression of long non-coding RNA (lncRNA) is closely related to disease progression. However, the role and mechanism of lncRNA H19 (lncH19) in sepsis-induced lung injury remain to be elucidated. METHODS Cercal ligation and puncture (CLP) mice models and lipopolysaccharide (LPS)-induced cell injury model were used to construct sepsis-induced lung injury in vivo and in vitro. The expression of lncH19, microRNA (miR)-301a-3p and adenylate cyclase 1 (Adcy1) mRNA was assessed using quantitative real-time PCR. The concentrations of inflammatory factors were determined by ELISA assay. Cell proliferation and apoptosis were determined using cell counting kit 8 assay, EdU staining and flow cytometry. The protein expression of apoptosis markers and Adcy1 was examined by western blot analysis. Oxidative stress was assessed by detecting the contents of oxidative stress markers. The interaction between miR-301a-3p and lncH19 or Adcy1 was confirmed using RNA pull-down assay, dual-luciferase reporter assay and RIP assay. RESULTS LncH19 was lowly expressed in CLP mice models and LPS-induced cell injury models. Overexpressed lncH19 could alleviate CLP-induced lung injury in mice, as well as LPS-induced cell apoptosis, inflammation and oxidative stress. MiR-301a-3p could be sponged by lncH19, and its overexpression could reverse the inhibition of lncH19 on LPS-induced cell injury. Adcy1 was a target of miR-301a-3p, and its expression was upregulated by lncH19. Silencing of Adcy1 could abolish the suppressive effect of miR-301a-3p inhibitor on LPS-induced cell injury. CONCLUSION LncH19 might inhibit sepsis-induced lung injury through acting as a sponge of miR-301a-3p to upregulate Adcy1.Highlights:LncH19 overexpression relieves CLP-induced lung injury and LPS-induced cell injury.LncH19 directly sponges miR-301a-3p.MiR-301a-3p targets Adcy1.
Collapse
Affiliation(s)
- Jingjing Hou
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Mei He
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Qiang Chen
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Siwei Liang
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| |
Collapse
|
13
|
Zhao YF. Free fatty acid receptors in the endocrine regulation of glucose metabolism: Insight from gastrointestinal-pancreatic-adipose interactions. Front Endocrinol (Lausanne) 2022; 13:956277. [PMID: 36246919 PMCID: PMC9554507 DOI: 10.3389/fendo.2022.956277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 09/14/2022] [Indexed: 11/25/2022] Open
Abstract
Glucose metabolism is primarily controlled by pancreatic hormones, with the coordinated assistance of the hormones from gastrointestine and adipose tissue. Studies have unfolded a sophisticated hormonal gastrointestinal-pancreatic-adipose interaction network, which essentially maintains glucose homeostasis in response to the changes in substrates and nutrients. Free fatty acids (FFAs) are the important substrates that are involved in glucose metabolism. FFAs are able to activate the G-protein coupled membrane receptors including GPR40, GPR120, GPR41 and GPR43, which are specifically expressed in pancreatic islet cells, enteroendocrine cells as well as adipocytes. The activation of FFA receptors regulates the secretion of hormones from pancreas, gastrointestine and adipose tissue to influence glucose metabolism. This review presents the effects of the FFA receptors on glucose metabolism via the hormonal gastrointestinal-pancreatic-adipose interactions and the underlying intracellular mechanisms. Furthermore, the development of therapeutic drugs targeting FFA receptors for the treatment of abnormal glucose metabolism such as type 2 diabetes mellitus is summarized.
Collapse
|
14
|
Gledhill LJ, Babey AM. Synthesis of the Mechanisms of Opioid Tolerance: Do We Still Say NO? Cell Mol Neurobiol 2021; 41:927-948. [PMID: 33704603 PMCID: PMC11448615 DOI: 10.1007/s10571-021-01065-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 02/12/2021] [Indexed: 10/21/2022]
Abstract
The use of morphine as a first-line agent for moderate-to-severe pain is limited by the development of analgesic tolerance. Initially opioid receptor desensitization in response to repeated stimulation, thought to underpin the establishment of tolerance, was linked to a compensatory increase in adenylate cyclase responsiveness. The subsequent demonstration of cross-talk between N-methyl-D-aspartate (NMDA) glutamate receptors and opioid receptors led to the recognition of a role for nitric oxide (NO), wherein blockade of NO synthesis could prevent tolerance developing. Investigations of the link between NO levels and opioid receptor desensitization implicated a number of events including kinase recruitment and peroxynitrite-mediated protein regulation. Recent experimental advances and the identification of new cellular constituents have expanded the potential signaling candidates to include unexpected, intermediary compounds not previously linked to this process such as zinc, histidine triad nucleotide-binding protein 1 (HINT1), micro-ribonucleic acid (mi-RNA) and regulator of G protein signaling Z (RGSZ). A further complication is a lack of consistency in the protocols used to create tolerance, with some using acute methods measured in minutes to hours and others using days. There is also an emphasis on the cellular changes that are extant only after tolerance has been established. Although a review of the literature demonstrates a lack of spatio-temporal detail, there still appears to be a pivotal role for nitric oxide, as well as both intracellular and intercellular cross-talk. The use of more consistent approaches to verify these underlying mechanism(s) could provide an avenue for targeted drug development to rescue opioid efficacy.
Collapse
Affiliation(s)
- Laura J Gledhill
- CURA Pharmacy, St. John of God Hospital, Bendigo, VIC, 3550, Australia
| | - Anna-Marie Babey
- Faculty of Medicine and Health, University of New England, Armidale, NSW, 2351, Australia.
| |
Collapse
|
15
|
Transcription factors regulated by cAMP in smooth muscle of the myometrium at human parturition. Biochem Soc Trans 2021; 49:997-1011. [PMID: 33860781 PMCID: PMC8106496 DOI: 10.1042/bst20201173] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 12/11/2022]
Abstract
Cyclic adenosine monophosphate (cAMP) contributes to maintenance of a quiescent (relaxed) state in the myometrium (i.e. uterine smooth muscle) during pregnancy, which most commonly has been attributed to activation of protein kinase A (PKA). PKA-mediated phosphorylation of cytosolic contractile apparatus components in myometrial smooth muscle cells (mSMCs) are known to promote relaxation. Additionally, PKA also regulates nuclear transcription factor (TF) activity to control expression of genes important to the labour process; these are mostly involved in actin-myosin interactions, cell-to-cell connectivity and inflammation, all of which influence mSMC transition from a quiescent to a contractile (pro-labour) phenotype. This review focuses on the evidence that cAMP modulates the activity of TFs linked to pro-labour gene expression, predominantly cAMP response element (CRE) binding TFs, nuclear factor κB (NF-κB), activator protein 1 (AP-1) family and progesterone receptors (PRs). This review also considers the more recently described exchange protein directly activated by cAMP (EPAC) that may oppose the pro-quiescent effects of PKA, as well as explores findings from other cell types that have the potential to be of novel relevance to cAMP action on TF function in the myometrium.
Collapse
|
16
|
Kim N, Shin S, Bae SW. cAMP Biosensors Based on Genetically Encoded Fluorescent/Luminescent Proteins. BIOSENSORS-BASEL 2021; 11:bios11020039. [PMID: 33572585 PMCID: PMC7911721 DOI: 10.3390/bios11020039] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/22/2021] [Accepted: 01/27/2021] [Indexed: 02/06/2023]
Abstract
Cyclic adenosine monophosphate (cAMP) plays a key role in signal transduction pathways as a second messenger. Studies on the cAMP dynamics provided useful scientific insights for drug development and treatment of cAMP-related diseases such as some cancers and prefrontal cortex disorders. For example, modulation of cAMP-mediated intracellular signaling pathways by anti-tumor drugs could reduce tumor growth. However, most early stage tools used for measuring the cAMP level in living organisms require cell disruption, which is not appropriate for live cell imaging or animal imaging. Thus, in the last decades, tools were developed for real-time monitoring of cAMP distribution or signaling dynamics in a non-invasive manner. Genetically-encoded sensors based on fluorescent proteins and luciferases could be powerful tools to overcome these drawbacks. In this review, we discuss the recent genetically-encoded cAMP sensors advances, based on single fluorescent protein (FP), Föster resonance energy transfer (FRET), single luciferase, and bioluminescence resonance energy transfer (BRET) for real-time non-invasive imaging.
Collapse
Affiliation(s)
- Namdoo Kim
- Department of Chemistry, Kongju National University, Gongju 32588, Korea;
| | - Seunghan Shin
- Green Chemistry & Materials Group, Korea Institute of Industrial Technology (KITECH), Cheonan 31056, Korea;
| | - Se Won Bae
- Green Chemistry & Materials Group, Korea Institute of Industrial Technology (KITECH), Cheonan 31056, Korea;
- Department of Chemistry and Cosmetics, Jeju National University, Jeju 63243, Korea
- Correspondence: ; Tel.: +82-64-754-3543
| |
Collapse
|
17
|
Sharma R. Network-based approach highlighting interplay among anti-hypertensives: target coding-genes: diseases. Sci Rep 2020; 10:20152. [PMID: 33214616 PMCID: PMC7677320 DOI: 10.1038/s41598-020-76605-1] [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: 03/15/2020] [Accepted: 08/10/2020] [Indexed: 11/22/2022] Open
Abstract
Elucidating the relation between the medicines: targets, targets: diseases and diseases: diseases are of fundamental significance as-is for societal benefit. Hypertension is one of the dangerous health conditions prevalent in society, is a risk factor for several other diseases if left untreated and anti-hypertensives (AHs) are the approved drugs to treat it. The goal of the study is to decipher the connection between hypertension with other health conditions, however, is challenging due to the large interactome. To fulfill the aim, the strategy involves prior clustering of the AHs into groups as per our previous method, followed by the analyzing functional association of the target coding-genes (tc-genes) and health conditions for each group. Following our recently published work where the AHs are clustered into six groups such that molecules having similar patterns come together, here, the distribution of molecular functions and the cellular components adopted by the tc-genes of each group are analyzed. The analyses indicate that kidney, heart, brain or lung related ailments are commonly associated with the tc-genes. The association of selective tc-genes to health conditions suggests a preference for certain health conditions despite many possibilities. Analyses of experimentally validated drug–drug combinations indicate the trend in successful AHs combinations. Clinically validated combinations bind different targets. Our study provides a promising methodology in a network-based approach that considers the influence of structural diversity of AHs to the functional perspective of tc-genes concerning the health conditions. The method could be extended to explore disease–disease relationships.
Collapse
Affiliation(s)
- Reetu Sharma
- Centre for Molecular Modeling, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, India.
| |
Collapse
|
18
|
Fox AP, Wagner KR, Towle VL, Xie KG, Xie Z. Caffeine reverses the unconsciousness produced by light anesthesia in the continued presence of isoflurane in rats. PLoS One 2020; 15:e0241818. [PMID: 33152041 PMCID: PMC7643991 DOI: 10.1371/journal.pone.0241818] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 10/20/2020] [Indexed: 12/17/2022] Open
Abstract
Currently no drugs are employed clinically to reverse the unconsciousness induced by general anesthetics. Our previous studies showed that caffeine, when given near the end of an anesthesia session, accelerated emergence from isoflurane anesthesia, likely caused by caffeine’s ability to elevate intracellular cAMP levels and to block adenosine receptors. These earlier studies showed that caffeine did not rouse either rats or humans from deep anesthesia (≥ 1 minimum alveolar concentration, MAC). In this current crossover study, we examined whether caffeine reversed the unconsciousness produced by light anesthesia (< 1 MAC) in the continued presence of isoflurane. The primary endpoint of this study was to measure isoflurane levels at the time of recovery of righting reflex, which was a proxy for consciousness. Rats were deeply anesthetized with 2% isoflurane (~1.5 MAC) for 20 minutes. Subsequently, isoflurane was reduced to 1.2% for 10 minutes, then by 0.2% every 10 min; animals were monitored until the recovery of righting reflex occurred, in the continued presence of isoflurane. Respiration rate, heart rate and electroencephalogram (EEG) were monitored. Our results show that caffeine-treated rats recovered their righting reflex at a significantly higher inspired isoflurane concentration, corresponding to light anesthesia, than the same rats treated with saline (control). Respiration rate and heart rate increased initially after caffeine injection but were then unchanged for the rest of the anesthesia session. Deep anesthesia is correlated with burst suppression in EEG recordings. Our data showed that caffeine transiently reduced the burst suppression time produced by deep anesthesia, suggesting that caffeine altered neuronal circuit function but not to a point where it caused arousal. In contrast, under light anesthesia, caffeine shifted the EEG power to high frequency beta and gamma bands. These data suggest that caffeine may represent a clinically viable drug to reverse the unconsciousness produced by light anesthesia.
Collapse
Affiliation(s)
- Aaron P. Fox
- Department of Neurobiology, Pharmacology and Physiology, The University of Chicago, Chicago, Illinois, United States of America
| | - Kyle R. Wagner
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, Illinois, United States of America
| | - Vernon L. Towle
- Department of Neurology, The University of Chicago, Chicago, Illinois, United States of America
| | - Kelvin G. Xie
- Department of Neurobiology, Pharmacology and Physiology, The University of Chicago, Chicago, Illinois, United States of America
| | - Zheng Xie
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
19
|
Almutairi F, Lee JK, Rada B. Regulator of G protein signaling 10: Structure, expression and functions in cellular physiology and diseases. Cell Signal 2020; 75:109765. [PMID: 32882407 PMCID: PMC7579743 DOI: 10.1016/j.cellsig.2020.109765] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 01/22/2023]
Abstract
Regulator of G protein signaling 10 (RGS10) belongs to the superfamily of RGS proteins, defined by the presence of a conserved RGS domain that canonically binds and deactivates heterotrimeric G-proteins. RGS proteins act as GTPase activating proteins (GAPs), which accelerate GTP hydrolysis on the G-protein α subunits and result in termination of signaling pathways downstream of G protein-coupled receptors. RGS10 is the smallest protein of the D/R12 subfamily and selectively interacts with Gαi proteins. It is widely expressed in many cells and tissues, with the highest expression found in the brain and immune cells. RGS10 expression is transcriptionally regulated via epigenetic mechanisms. Although RGS10 lacks multiple of the defined regulatory domains found in other RGS proteins, RGS10 contains post-translational modification sites regulating its expression, localization, and function. Additionally, RGS10 is a critical protein in the regulation of physiological processes in multiple cells, where dysregulation of its expression has been implicated in various diseases including Parkinson's disease, multiple sclerosis, osteopetrosis, chemoresistant ovarian cancer and cardiac hypertrophy. This review summarizes RGS10 features and its regulatory mechanisms, and discusses the known functions of RGS10 in cellular physiology and pathogenesis of several diseases.
Collapse
Affiliation(s)
- Faris Almutairi
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA; Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Jae-Kyung Lee
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Balázs Rada
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA.
| |
Collapse
|
20
|
Mechanisms and Regulation of Neuronal GABA B Receptor-Dependent Signaling. Curr Top Behav Neurosci 2020; 52:39-79. [PMID: 32808092 DOI: 10.1007/7854_2020_129] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
γ-Aminobutyric acid B receptors (GABABRs) are broadly expressed throughout the central nervous system where they play an important role in regulating neuronal excitability and synaptic transmission. GABABRs are G protein-coupled receptors that mediate slow and sustained inhibitory actions via modulation of several downstream effector enzymes and ion channels. GABABRs are obligate heterodimers that associate with diverse arrays of proteins to form modular complexes that carry out distinct physiological functions. GABABR-dependent signaling is fine-tuned and regulated through a multitude of mechanisms that are relevant to physiological and pathophysiological states. This review summarizes the current knowledge on GABABR signal transduction and discusses key factors that influence the strength and sensitivity of GABABR-dependent signaling in neurons.
Collapse
|
21
|
van der Horst J, Greenwood IA, Jepps TA. Cyclic AMP-Dependent Regulation of Kv7 Voltage-Gated Potassium Channels. Front Physiol 2020; 11:727. [PMID: 32695022 PMCID: PMC7338754 DOI: 10.3389/fphys.2020.00727] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/04/2020] [Indexed: 01/08/2023] Open
Abstract
Voltage-gated Kv7 potassium channels, encoded by KCNQ genes, have major physiological impacts cardiac myocytes, neurons, epithelial cells, and smooth muscle cells. Cyclic adenosine monophosphate (cAMP), a well-known intracellular secondary messenger, can activate numerous downstream effector proteins, generating downstream signaling pathways that regulate many functions in cells. A role for cAMP in ion channel regulation has been established, and recent findings show that cAMP signaling plays a role in Kv7 channel regulation. Although cAMP signaling is recognized to regulate Kv7 channels, the precise molecular mechanism behind the cAMP-dependent regulation of Kv7 channels is complex. This review will summarize recent research findings that support the mechanisms of cAMP-dependent regulation of Kv7 channels.
Collapse
Affiliation(s)
- Jennifer van der Horst
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Iain A Greenwood
- Molecular and Clinical Sciences Institute, St. George's University of London, London, United Kingdom
| | - Thomas A Jepps
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Ayukawa K, Suzuki C, Ogasawara H, Kinoshita T, Furuno M, Suzuki G. Development of a High-Throughput Screening-Compatible Assay for Discovery of GPR3 Inverse Agonists Using a cAMP Biosensor. SLAS DISCOVERY 2019; 25:287-298. [PMID: 31516076 DOI: 10.1177/2472555219875101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
While G-protein-coupled receptors (GPCRs) represent the largest class of cell surface proteins, there are ≥100 orphan GPCRs whose endogenous ligands are unknown. Accordingly, these could prove to be potential therapeutic targets for the pharmaceutical intervention of various diseases. Constitutively active orphan GPCRs are activated without ligands; thus, inverse agonists may be very useful pharmacological tools for inhibiting constitutive activity. However, in general, inverse agonist screening is considered more difficult to perform with high quality than antagonist screening, particularly due to the narrow assay window. We developed a high-throughput screening (HTS)-compatible assay to identify inverse agonists of GPR3. GPR3 is expressed in the central nervous system (CNS) and is known to be related to Alzheimer's disease and other CNS diseases. The GPR3 inducible cell line was established using T-REx 293 cells that stably expressed the tetracycline repressor protein, and the cAMP biosensor, GloSensor, was stably co-expressed. After optimization of the induction level of GPR3 and assay conditions, the GloSensor assay showed an approximately 20-fold signal-to-background ratio and high sensitivity. Using the HTS method, we successfully screened a library of hundreds of thousands of compounds for the inhibition of constitutive activity with good quality and excellent reproducibility. Finally, 35 compounds were identified as GPR3 selective inverse agonists. This inverse agonist screening approach using GloSensor in combination with the inducible expression of orphan GPCR indicates universal applicability to the search for inverse agonists of constitutively active orphan GPCRs.
Collapse
Affiliation(s)
- Kumiko Ayukawa
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Takatsuki, Osaka, Japan
| | - Chie Suzuki
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Takatsuki, Osaka, Japan
| | - Hiroyuki Ogasawara
- Pharmaceutical Frontier Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Kanazawa-ku, Yokohama, Kanagawa, Japan
| | - Tomomi Kinoshita
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Takatsuki, Osaka, Japan
| | - Masahiro Furuno
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Takatsuki, Osaka, Japan
| | - Gentaroh Suzuki
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Takatsuki, Osaka, Japan
| |
Collapse
|
23
|
Zhang H, Nielsen AL, Strømgaard K. Recent achievements in developing selective Gqinhibitors. Med Res Rev 2019; 40:135-157. [DOI: 10.1002/med.21598] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/09/2019] [Accepted: 04/26/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Hang Zhang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Co‐innovation Center of Henan Province for New Drug R&D and Preclinical Safety, and School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou Henan China
| | - Alexander L. Nielsen
- Department of Drug Design and Pharmacology, Center for BiopharmaceuticalsUniversity of CopenhagenCopenhagen Denmark
| | - Kristian Strømgaard
- Department of Drug Design and Pharmacology, Center for BiopharmaceuticalsUniversity of CopenhagenCopenhagen Denmark
| |
Collapse
|
24
|
Iring A, Jin YJ, Albarrán-Juárez J, Siragusa M, Wang S, Dancs PT, Nakayama A, Tonack S, Chen M, Künne C, Sokol AM, Günther S, Martínez A, Fleming I, Wettschureck N, Graumann J, Weinstein LS, Offermanns S. Shear stress-induced endothelial adrenomedullin signaling regulates vascular tone and blood pressure. J Clin Invest 2019; 129:2775-2791. [PMID: 31205027 DOI: 10.1172/jci123825] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 05/01/2019] [Indexed: 12/22/2022] Open
Abstract
Hypertension is a primary risk factor for cardiovascular diseases including myocardial infarction and stroke. Major determinants of blood pressure are vasodilatory factors such as nitric oxide (NO) released from the endothelium under the influence of fluid shear stress exerted by the flowing blood. Several endothelial signaling processes mediating fluid shear stress-induced formation and release of vasodilatory factors have been described. It is, however, still poorly understood how fluid shear stress induces these endothelial responses. Here we show that the endothelial mechanosensitive cation channel PIEZO1 mediated fluid shear stress-induced release of adrenomedullin, which in turn activated its Gs-coupled receptor. The subsequent increase in cAMP levels promoted the phosphorylation of endothelial NO synthase (eNOS) at serine 633 through protein kinase A (PKA), leading to the activation of the enzyme. This Gs/PKA-mediated pathway synergized with the AKT-mediated pathways leading to eNOS phosphorylation at serine 1177. Mice with endothelium-specific deficiency of adrenomedullin, the adrenomedullin receptor, or Gαs showed reduced flow-induced eNOS activation and vasodilation and developed hypertension. Our data identify fluid shear stress-induced PIEZO1 activation as a central regulator of endothelial adrenomedullin release and establish the adrenomedullin receptor and subsequent Gs-mediated formation of cAMP as a critical endothelial mechanosignaling pathway regulating basal endothelial NO formation, vascular tone, and blood pressure.
Collapse
Affiliation(s)
- Andras Iring
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Young-June Jin
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Julián Albarrán-Juárez
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Mauro Siragusa
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany
| | - ShengPeng Wang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Yanta District, Xi'an, China
| | - Péter T Dancs
- Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - Akiko Nakayama
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Sarah Tonack
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Min Chen
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | | | - Anna M Sokol
- Scientific Service Group Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | - Alfredo Martínez
- Oncology Area, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Centre for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany
| | - Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany.,Centre for Molecular Medicine, Medical Faculty, Goethe University, Frankfurt am Main, Germany
| | - Johannes Graumann
- German Centre for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany.,Scientific Service Group Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Lee S Weinstein
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany.,Centre for Molecular Medicine, Medical Faculty, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
25
|
Urizar-Arenaza I, Osinalde N, Akimov V, Puglia M, Candenas L, Pinto FM, Muñoa-Hoyos I, Gianzo M, Matorras R, Irazusta J, Blagoev B, Subiran N, Kratchmarova I. Phosphoproteomic and Functional Analyses Reveal Sperm-specific Protein Changes Downstream of Kappa Opioid Receptor in Human Spermatozoa. Mol Cell Proteomics 2019; 18:S118-S131. [PMID: 30622161 PMCID: PMC6427232 DOI: 10.1074/mcp.ra118.001133] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/27/2018] [Indexed: 12/11/2022] Open
Abstract
G-protein coupled receptors (GPCRs) belong to the seven transmembrane receptor superfamily that transduce signals via G proteins in response to external stimuli to initiate different intracellular signaling pathways which culminate in specific cellular responses. The expression of diverse GPCRs at the plasma membrane of human spermatozoa suggests their involvement in the regulation of sperm fertility. However, the signaling events downstream of many GPCRs in spermatozoa remain uncharacterized. Here, we selected the kappa-opioid receptor (KOR) as a study model and applied phosphoproteomic approach based on TMT labeling and LC-MS/MS analyses. Quantitative coverage of more than 5000 proteins with over 3500 phosphorylation sites revealed changes in the phosphorylation levels of sperm-specific proteins involved in the regulation of the sperm fertility in response to a specific agonist of KOR, U50488H. Further functional studies indicate that KOR could be involved in the regulation of sperm fertile capacity by modulation of calcium channels. Our findings suggest that human spermatozoa possess unique features in the molecular mechanisms downstream of GPCRs which could be key regulators of sperm fertility and improved knowledge of these specific processes may contribute to the development of useful biochemical tools for diagnosis and treatment of male infertility.
Collapse
Affiliation(s)
- Itziar Urizar-Arenaza
- From the ‡Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain, 49840;; Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain, 48903
| | - Nerea Osinalde
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Araba, Spain, 01006
| | - Vyacheslav Akimov
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark, 5320
| | - Michele Puglia
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark, 5320
| | - Luz Candenas
- Instituto de Investigaciones Químicas, CSIC, Sevilla, Spain, 41092
| | | | - Iraia Muñoa-Hoyos
- From the ‡Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain, 49840;; Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain, 48903
| | - Marta Gianzo
- From the ‡Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain, 49840;; Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain, 48903
| | - Roberto Matorras
- Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain, 48903
| | - Jon Irazusta
- From the ‡Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain, 49840
| | - Blagoy Blagoev
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark, 5320
| | - Nerea Subiran
- From the ‡Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain, 49840;; Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain, 48903;.
| | - Irina Kratchmarova
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark, 5320;.
| |
Collapse
|
26
|
Touhara KK, MacKinnon R. Molecular basis of signaling specificity between GIRK channels and GPCRs. eLife 2018; 7:42908. [PMID: 30526853 PMCID: PMC6335053 DOI: 10.7554/elife.42908] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 12/08/2018] [Indexed: 12/18/2022] Open
Abstract
Stimulated muscarinic acetylcholine receptors (M2Rs) release Gβγ subunits, which slow heart rate by activating a G protein-gated K+ channel (GIRK). Stimulated β2 adrenergic receptors (β2ARs) also release Gβγ subunits, but GIRK is not activated. This study addresses the mechanism underlying this specificity of GIRK activation by M2Rs. K+ currents and bioluminescence resonance energy transfer between labelled G proteins and GIRK show that M2Rs catalyze Gβγ subunit release at higher rates than β2ARs, generating higher Gβγ concentrations that activate GIRK and regulate other targets of Gβγ. The higher rate of Gβγ release is attributable to a faster G protein coupled receptor – G protein trimer association rate in M2R compared to β2AR. Thus, a rate difference in a single kinetic step accounts for specificity.
Collapse
Affiliation(s)
- Kouki K Touhara
- Laboratory of Molecular Neurobiology and Biophysics, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Roderick MacKinnon
- Laboratory of Molecular Neurobiology and Biophysics, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| |
Collapse
|
27
|
Soto-Velasquez M, Hayes MP, Alpsoy A, Dykhuizen EC, Watts VJ. A Novel CRISPR/Cas9-Based Cellular Model to Explore Adenylyl Cyclase and cAMP Signaling. Mol Pharmacol 2018; 94:963-972. [PMID: 29950405 PMCID: PMC6064782 DOI: 10.1124/mol.118.111849] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/20/2018] [Indexed: 11/22/2022] Open
Abstract
Functional characterization of adenylyl cyclase (AC) isoforms has proven challenging in mammalian cells because of the endogenous expression of multiple AC isoforms and the high background cAMP levels induced by nonselective AC activators. To simplify the characterization of individual transmembrane AC (mAC) isoforms, we generated a human embryonic kidney cell line 293 (HEK293) with low cAMP levels by knocking out two highly expressed ACs, AC3 and AC6, using CRISPR/Cas9 technology. Stable HEK293 cell lines lacking either AC6 (HEK-ACΔ6) or both AC3 and AC6 (HEK-ACΔ3/6) were generated. Knockout was confirmed genetically and by comparing cAMP responses of the knockout cells to the parental cell line. HEK-ACΔ6 and HEK-ACΔ3/6 cells revealed an 85% and 95% reduction in the forskolin-stimulated cAMP response, respectively. Forskolin- and Gαs-coupled receptor-induced activation was examined for the nine recombinant mAC isoforms in the HEK-ACΔ3/6 cells. Forskolin-mediated cAMP accumulation for AC1-6 and AC8 revealed 10- to 250-fold increases over the basal cAMP levels. All nine mAC isoforms, except AC8, also exhibited significantly higher cAMP levels than the control cells after Gαs-coupled receptor activation. Isoform-specific AC regulation by protein kinases and Ca2+/calmodulin was also recapitulated in the knockout cells. Furthermore, the utility of the HEK-ACΔ3/6 cell line was demonstrated by characterizing the activity of novel AC1 forskolin binding-site mutants. Hence, we have developed a HEK293 cell line deficient of endogenous AC3 and AC6 with low cAMP background levels for studies of cAMP signaling and AC isoform regulation.
Collapse
Affiliation(s)
- Monica Soto-Velasquez
- Department of Medicinal Chemistry and Molecular Pharmacology (M.S.-V., M.P.H., A.A., E.C.D., V.J.W.), Purdue Institute of Drug Discovery (E.C.D., V.J.W.), Purdue University, West Lafayette, Indiana
| | - Michael P Hayes
- Department of Medicinal Chemistry and Molecular Pharmacology (M.S.-V., M.P.H., A.A., E.C.D., V.J.W.), Purdue Institute of Drug Discovery (E.C.D., V.J.W.), Purdue University, West Lafayette, Indiana
| | - Aktan Alpsoy
- Department of Medicinal Chemistry and Molecular Pharmacology (M.S.-V., M.P.H., A.A., E.C.D., V.J.W.), Purdue Institute of Drug Discovery (E.C.D., V.J.W.), Purdue University, West Lafayette, Indiana
| | - Emily C Dykhuizen
- Department of Medicinal Chemistry and Molecular Pharmacology (M.S.-V., M.P.H., A.A., E.C.D., V.J.W.), Purdue Institute of Drug Discovery (E.C.D., V.J.W.), Purdue University, West Lafayette, Indiana
| | - Val J Watts
- Department of Medicinal Chemistry and Molecular Pharmacology (M.S.-V., M.P.H., A.A., E.C.D., V.J.W.), Purdue Institute of Drug Discovery (E.C.D., V.J.W.), Purdue University, West Lafayette, Indiana
| |
Collapse
|
28
|
TERUNUMA M. Diversity of structure and function of GABA B receptors: a complexity of GABA B-mediated signaling. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2018; 94:390-411. [PMID: 30541966 PMCID: PMC6374141 DOI: 10.2183/pjab.94.026] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 10/09/2018] [Indexed: 05/24/2023]
Abstract
γ-aminobutyric acid type B (GABAB) receptors are broadly expressed in the nervous system and play an important role in neuronal excitability. GABAB receptors are G protein-coupled receptors that mediate slow and prolonged inhibitory action, via activation of Gαi/o-type proteins. GABAB receptors mediate their inhibitory action through activating inwardly rectifying K+ channels, inactivating voltage-gated Ca2+ channels, and inhibiting adenylate cyclase. Functional GABAB receptors are obligate heterodimers formed by the co-assembly of R1 and R2 subunits. It is well established that GABAB receptors interact not only with G proteins and effectors but also with various proteins. This review summarizes the structure, subunit isoforms, and function of GABAB receptors, and discusses the complexity of GABAB receptors, including how receptors are localized in specific subcellular compartments, the mechanism regulating cell surface expression and mobility of the receptors, and the diversity of receptor signaling through receptor crosstalk and interacting proteins.
Collapse
Affiliation(s)
- Miho TERUNUMA
- Division of Oral Biochemistry, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
29
|
Grzelka K, Kurowski P, Gawlak M, Szulczyk P. Noradrenaline Modulates the Membrane Potential and Holding Current of Medial Prefrontal Cortex Pyramidal Neurons via β 1-Adrenergic Receptors and HCN Channels. Front Cell Neurosci 2017; 11:341. [PMID: 29209170 PMCID: PMC5701640 DOI: 10.3389/fncel.2017.00341] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/16/2017] [Indexed: 12/27/2022] Open
Abstract
The medial prefrontal cortex (mPFC) receives dense noradrenergic projections from the locus coeruleus. Adrenergic innervation of mPFC pyramidal neurons plays an essential role in both physiology (control of memory formation, attention, working memory, and cognitive behavior) and pathophysiology (attention deficit hyperactivity disorder, posttraumatic stress disorder, cognitive deterioration after traumatic brain injury, behavioral changes related to addiction, Alzheimer's disease and depression). The aim of this study was to elucidate the mechanism responsible for adrenergic receptor-mediated control of the resting membrane potential in layer V mPFC pyramidal neurons. The membrane potential or holding current of synaptically isolated layer V mPFC pyramidal neurons was recorded in perforated-patch and classical whole-cell configurations in slices from young rats. Application of noradrenaline (NA), a neurotransmitter with affinity for all types of adrenergic receptors, evoked depolarization or inward current in the tested neurons irrespective of whether the recordings were performed in the perforated-patch or classical whole-cell configuration. The effect of noradrenaline depended on β1- and not α1- or α2-adrenergic receptor stimulation. Activation of β1-adrenergic receptors led to an increase in inward Na+ current through hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, which carry a mixed Na+/K+ current. The protein kinase A- and C-, glycogen synthase kinase-3β- and tyrosine kinase-linked signaling pathways were not involved in the signal transduction between β1-adrenergic receptors and HCN channels. The transduction system operated in a membrane-delimited fashion and involved the βγ subunit of G-protein. Thus, noradrenaline controls the resting membrane potential and holding current in mPFC pyramidal neurons through β1-adrenergic receptors, which in turn activate HCN channels via a signaling pathway involving the βγ subunit.
Collapse
Affiliation(s)
- Katarzyna Grzelka
- Laboratory of Physiology and Pathophysiology, Centre for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
| | | | | | - Paweł Szulczyk
- Laboratory of Physiology and Pathophysiology, Centre for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
30
|
Khammy MM, Dalsgaard T, Larsen PH, Christoffersen CT, Clausen D, Rasmussen LK, Folkersen L, Grunnet M, Kehler J, Aalkjaer C, Nielsen J. PDE1A inhibition elicits cGMP-dependent relaxation of rat mesenteric arteries. Br J Pharmacol 2017; 174:4186-4198. [PMID: 28910498 DOI: 10.1111/bph.14034] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 08/18/2017] [Accepted: 09/07/2017] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND AND PURPOSE PDE1, a subfamily of cyclic nucleotide PDEs consisting of three isoforms, PDE1A, PDE1B and PDE1C, has been implicated in the regulation of vascular tone. The PDE1 isoform(s) responsible for tone regulation is unknown. This study used isoform-preferring PDE1 inhibitors, Lu AF58027, Lu AF64196, Lu AF66896 and Lu AF67897, to investigate the relative contribution of PDE1 isoforms to regulation of vascular tone. EXPERIMENTAL APPROACH In rat mesenteric arteries, expression and localization of Pde1 isoforms were determined by quantitative PCR and in situ hybridization, and physiological impact of PDE1 inhibition was evaluated by isometric tension recordings. KEY RESULTS In rat mesenteric arteries, Pde1a mRNA expression was higher than Pde1b and Pde1c. In situ hybridization revealed localization of Pde1a to vascular smooth muscle cells (VSMCs) and only minor appearance of Pde1b and Pde1c. The potency of the PDE1 inhibitors at eliciting relaxation showed excellent correlation with their potency at inhibiting PDE1A. Thus, Lu AF58027 was the most potent at inhibiting PDE1A and was also the most potent at eliciting relaxation in mesenteric arteries. Inhibition of NOS with l-NAME, soluble GC with ODQ or PKG with Rp-8-Br-PET-cGMP all attenuated the inhibitory effect of PDE1 on relaxation, whereas PKA inhibition with H89 had no effect. CONCLUSIONS AND IMPLICATIONS Pde1a is the dominant PDE1 isoform present in VSMCs, and relaxation mediated by PDE1A inhibition is predominantly driven by enhanced cGMP signalling. These results imply that isoform-selective PDE1 inhibitors are powerful investigative tools allowing examination of physiological and pathological roles of PDE1 isoforms.
Collapse
Affiliation(s)
- Makhala Michell Khammy
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Thomas Dalsgaard
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Dorte Clausen
- Division of Synaptic Transmission, H. Lundbeck A/S, Valby, Denmark
| | | | - Lasse Folkersen
- Division of Synaptic Transmission, H. Lundbeck A/S, Valby, Denmark
| | - Morten Grunnet
- Division of Synaptic Transmission, H. Lundbeck A/S, Valby, Denmark
| | - Jan Kehler
- Division of Discovery Chemistry and DMPK, H. Lundbeck A/S, Valby, Denmark
| | - Christian Aalkjaer
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jacob Nielsen
- Division of Synaptic Transmission, H. Lundbeck A/S, Valby, Denmark
| |
Collapse
|
31
|
Fong R, Khokhar S, Chowdhury AN, Xie KG, Wong JHY, Fox AP, Xie Z. Caffeine accelerates recovery from general anesthesia via multiple pathways. J Neurophysiol 2017; 118:1591-1597. [PMID: 28659466 PMCID: PMC5596131 DOI: 10.1152/jn.00393.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 06/22/2017] [Accepted: 06/22/2017] [Indexed: 12/19/2022] Open
Abstract
Various studies have explored different ways to speed emergence from anesthesia. Previously, we have shown that three drugs that elevate intracellular cAMP (forskolin, theophylline, and caffeine) accelerate emergence from anesthesia in rats. However, our earlier studies left two main questions unanswered. First, were cAMP-elevating drugs effective at all anesthetic concentrations? Second, given that caffeine was the most effective of the drugs tested, why was caffeine more effective than forskolin since both drugs elevate cAMP? In our current study, emergence time from anesthesia was measured in adult rats exposed to 3% isoflurane for 60 min. Caffeine dramatically accelerated emergence from anesthesia, even at the high level of anesthetic employed. Caffeine has multiple actions including blockade of adenosine receptors. We show that the selective A2a adenosine receptor antagonist preladenant or the intracellular cAMP ([cAMP]i)-elevating drug forskolin, accelerated recovery from anesthesia. When preladenant and forskolin were tested together, the effect on anesthesia recovery time was additive indicating that these drugs operate via different pathways. Furthermore, the combination of preladenant and forskolin was about as effective as caffeine suggesting that both A2A receptor blockade and [cAMP]i elevation play a role in caffeine's ability to accelerate emergence from anesthesia. Because anesthesia in rodents is thought to be similar to that in humans, these results suggest that caffeine might allow for rapid and uniform emergence from general anesthesia in humans at all anesthetic concentrations and that both the elevation of [cAMP]i and adenosine receptor blockade play a role in this response.NEW & NOTEWORTHY Currently, there is no method to accelerate emergence from anesthesia. Patients "wake" when they clear the anesthetic from their systems. Previously, we have shown that caffeine can accelerate emergence from anesthesia. In this study, we show that caffeine is effective even at high levels of anesthetic. We also show that caffeine operates by both elevating intracellular cAMP levels and by blocking adenosine receptors. This complicated pharmacology makes caffeine especially effective in accelerating emergence from anesthesia.
Collapse
Affiliation(s)
- Robert Fong
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, Illinois
| | - Suhail Khokhar
- College of Medicine, University of Illinois, School of Life Sciences, Chicago, Illinois
| | - Atif N Chowdhury
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kelvin G Xie
- School of Engineering and Applied Science, Washington University, St Louis, Missouri
| | | | - Aaron P Fox
- Department of Neurobiology, Pharmacology and Physiology, The University of Chicago, Chicago, Illinois
| | - Zheng Xie
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, Illinois;
| |
Collapse
|
32
|
Kinetic properties and adrenergic control of TREK-2-like channels in rat medial prefrontal cortex (mPFC) pyramidal neurons. Brain Res 2017; 1665:95-104. [PMID: 28438532 DOI: 10.1016/j.brainres.2017.04.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/03/2017] [Accepted: 04/14/2017] [Indexed: 02/01/2023]
Abstract
TREK-2-like channels were identified on the basis of electrophysiological and pharmacological tests performed on freshly isolated and enzymatically/mechanically dispersed pyramidal neurons of the rat medial prefrontal cortex (mPFC). Single-channel currents were recorded in cell-attached configuration and the impact of adrenergic receptors (α1, α2, β) stimulation on spontaneously appearing TREK-2-like channel activity was tested. The obtained results indicate that noradrenaline decreases the mean open probability of TREK-2-like channel currents by activation of β1 but not of α1- and α2-adrenergic receptors. Mean open time and channel conductance were not affected. The system of intracellular signaling pathways depends on the activation of protein kinase A. We also show that adrenergic control of TREK-2-like channel currents by adrenergic receptors was similar in pyramidal neurons isolated from young, adolescent, and adult rats. Immunofluorescent confocal scans of mPFC slices confirmed the presence of the TREK-2 protein, which was abundant in layer V pyramidal neurons. The role of TREK-2-like channel control by adrenergic receptors is discussed.
Collapse
|
33
|
López-Cardona AP, Pérez-Cerezales S, Fernández-González R, Laguna-Barraza R, Pericuesta E, Agirregoitia N, Gutiérrez-Adán A, Agirregoitia E. CB 1 cannabinoid receptor drives oocyte maturation and embryo development via PI3K/Akt and MAPK pathways. FASEB J 2017; 31:3372-3382. [PMID: 28428264 DOI: 10.1096/fj.201601382rr] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 04/05/2017] [Indexed: 11/11/2022]
Abstract
Endocannabinoids have been recognized as mediators of practically all reproductive events in mammals. However, little is known about the role of this system in oocyte maturation. In a mouse model, we observed that activation of cannabinoid receptor 1 (CB1) during in vitro oocyte maturation modulated the phosphorylation status of Akt and ERK1/2 and enhanced the subsequent embryo production. In the absence of CB1, in vivo oocyte maturation was impaired and embryo development delayed. Cannabinoid receptor 2 (CB2) was unable to rescue these effects. Finally, we confirmed abnormal oocyte maturation rather than impaired embryonic transport through the oviduct in CB1 knockouts. Our data suggest that cannabinoid agonists may be useful in vitro maturation supplements. For in vitro fertilization patients intolerant to gonadotropins, this could be a promising and only option.-López-Cardona, A. P., Pérez-Cerezales, S., Fernández-González, R., Laguna-Barraza, R., Pericuesta, E., Agirregoitia, N., Gutiérrez-Adán, A., Agirregoitia, E. CB1 cannabinoid receptor drives oocyte maturation and embryo development via PI3K/Akt and MAPK pathways.
Collapse
Affiliation(s)
- Angela Patricia López-Cardona
- Department of Animal Reproduction, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain.,Groupo de Investigación (G.I.)-Biogénesis, Universidad de Antioquia, Medellín, Colombia
| | - Serafín Pérez-Cerezales
- Department of Animal Reproduction, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain
| | - Raúl Fernández-González
- Department of Animal Reproduction, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain
| | - Ricardo Laguna-Barraza
- Department of Animal Reproduction, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain
| | - Eva Pericuesta
- Department of Animal Reproduction, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain
| | - Naiara Agirregoitia
- Department of Physiology, Faculty of Medicine and Nursing University of the Basque Country (UPV/EHU), Leioa, Bizkaia
| | - Alfonso Gutiérrez-Adán
- Department of Animal Reproduction, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain
| | - Ekaitz Agirregoitia
- Department of Physiology, Faculty of Medicine and Nursing University of the Basque Country (UPV/EHU), Leioa, Bizkaia
| |
Collapse
|
34
|
Razzak MA, Hossain MS, Radzi ZB, Yahya NAB, Czernuszka J, Rahman MT. Cellular and Molecular Responses to Mechanical Expansion of Tissue. Front Physiol 2016; 7:540. [PMID: 27899897 PMCID: PMC5111402 DOI: 10.3389/fphys.2016.00540] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 10/27/2016] [Indexed: 01/08/2023] Open
Abstract
The increased use of tissue expander in the past decades and its potential market values in near future give enough reasons to sum up the consequences of tissue expansion. Furthermore, the patients have the right to know underlying mechanisms of adaptation of inserted biomimetic, its bioinspired materials and probable complications. The mechanical strains during tissue expansion are related to several biological phenomena. Tissue remodeling during the expansion is highly regulated and depends on the signal transduction. Any alteration may lead to tumor formation, necrosis and/or apoptosis. In this review, stretch induced cell proliferation, apoptosis, the roles of growth factors, stretch induced ion channels, and roles of second messengers are organized. It is expected that readers from any background can understand and make a decision about tissue expansion.
Collapse
Affiliation(s)
- Muhammad Abdur Razzak
- Department of Children's Dentistry and Orthodontics, Faculty of Dentistry, University of Malaya Kuala Lumpur, Malaysia
| | - Md Sanower Hossain
- Department of Children's Dentistry and Orthodontics, Faculty of Dentistry, University of Malaya Kuala Lumpur, Malaysia
| | - Zamri Bin Radzi
- Department of Children's Dentistry and Orthodontics, Faculty of Dentistry, University of Malaya Kuala Lumpur, Malaysia
| | - Noor Azlin B Yahya
- Department of Children's Dentistry and Orthodontics, Faculty of Dentistry, University of Malaya Kuala Lumpur, Malaysia
| | - Jan Czernuszka
- Department of Materials, University of Oxford Oxford, UK
| | - Mohammad T Rahman
- Department of Children's Dentistry and Orthodontics, Faculty of Dentistry, University of Malaya Kuala Lumpur, Malaysia
| |
Collapse
|
35
|
Hayes MP, Roman DL. Regulator of G Protein Signaling 17 as a Negative Modulator of GPCR Signaling in Multiple Human Cancers. AAPS JOURNAL 2016; 18:550-9. [PMID: 26928451 DOI: 10.1208/s12248-016-9894-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 02/15/2016] [Indexed: 02/08/2023]
Abstract
Regulators of G protein signaling (RGS) proteins modulate G protein-coupled receptor (GPCR) signaling networks by terminating signals produced by active Gα subunits. RGS17, a member of the RZ subfamily of RGS proteins, is typically only expressed in appreciable amounts in the human central nervous system, but previous works have shown that RGS17 expression is selectively upregulated in a number of malignancies, including lung, breast, prostate, and hepatocellular carcinoma. In addition, this upregulation of RGS17 is associated with a more aggressive cancer phenotype, as increased proliferation, migration, and invasion are observed. Conversely, decreased RGS17 expression diminishes the response of ovarian cancer cells to agents commonly used during chemotherapy. These somewhat contradictory roles of RGS17 in cancer highlight the need for selective, high-affinity inhibitors of RGS17 to use as chemical probes to further the understanding of RGS17 biology. Based on current evidence, these compounds could potentially have clinical utility as novel chemotherapeutics in the treatment of lung, prostate, breast, and liver cancers. Recent advances in screening technologies to identify potential inhibitors coupled with increasing knowledge of the structural requirements of RGS-Gα protein-protein interaction inhibitors make the future of drug discovery efforts targeting RGS17 promising. This review highlights recent findings related to RGS17 as both a canonical and atypical RGS protein, its role in various human disease states, and offers insights on small molecule inhibition of RGS17.
Collapse
Affiliation(s)
- Michael P Hayes
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, Iowa, USA
| | - David L Roman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, Iowa, USA. .,Cancer Signaling and Experimental Therapeutics Program, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA. .,, 115 S. Grand Avenue, S327 PHAR, Iowa City, Iowa, 52242, USA.
| |
Collapse
|
36
|
Li L, Matsuoka I, Sakamoto K, Kimura J. Differential effects of lysophosphatidylcholine and ACh on muscarinic K(+),non-selective cation and Ca(2+) currents in guinea-pig atrial cells. Fukushima J Med Sci 2016; 62:27-35. [PMID: 26911304 DOI: 10.5387/fms.2015-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We compared the effects of lysophosphatidylcholine (LPC) and acetylcholine (ACh) on IK(ACh), ICa and a non-selective cation current (INSC) in guinea-pig atrial myocytes to clarify whether LPC and ACh activate similar Gi/o-coupled effector systems. IK(ACh), ICa and INSC were analyzed in single atrial myocytes by the whole cell patch-clamp. LPC induced INSC in a concentration-dependent manner in atrial cells. ACh activated IK(ACh), but failed to evoke INSC. LPC also activated IK(ACh) but with significantly less potency than ACh. The effects of both ligands on IK(ACh) were inhibited by intracellular loading of pre-activated PTX. This treatment also inhibited LPC-induced INSC, indicating that IK(ACh) and INSC induced by LPC are both mediated by Gi/o. LPC and ACh had similar potencies in inhibiting ICa, which was pre-augmented by forskolin, indicating that LPC and ACh activate similar amounts of α-subunits of Gi/o. The different effects of LPC and ACh on IK(ACh) and INSC may suggest that LPC and ACh activate Gi/o having different types of βγ subunits, and that LPC-induced INSC may be mediated by βγ subunits of Gi/o, which are less effective in inducing IK(ACh).
Collapse
Affiliation(s)
- Libing Li
- Department of Pharmacology, School of Medicine, Fukushima Medical University
| | | | | | | |
Collapse
|
37
|
Frese S, Ruebner M, Suhr F, Konou TM, Tappe KA, Toigo M, Jung HH, Henke C, Steigleder R, Strissel PL, Huebner H, Beckmann MW, van der Keylen P, Schoser B, Schiffer T, Frese L, Bloch W, Strick R. Long-Term Endurance Exercise in Humans Stimulates Cell Fusion of Myoblasts along with Fusogenic Endogenous Retroviral Genes In Vivo. PLoS One 2015; 10:e0132099. [PMID: 26154387 PMCID: PMC4495930 DOI: 10.1371/journal.pone.0132099] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 06/10/2015] [Indexed: 11/21/2022] Open
Abstract
Myogenesis is defined as growth, differentiation and repair of muscles where cell fusion of myoblasts to multinucleated myofibers is one major characteristic. Other cell fusion events in humans are found with bone resorbing osteoclasts and placental syncytiotrophoblasts. No unifying gene regulation for natural cell fusions has been found. We analyzed skeletal muscle biopsies of competitive cyclists for muscle-specific attributes and expression of human endogenous retrovirus (ERV) envelope genes due to their involvement in cell fusion of osteoclasts and syncytiotrophoblasts. Comparing muscle biopsies from post- with the pre-competitive seasons a significant 2.25-fold increase of myonuclei/mm fiber, a 2.38-fold decrease of fiber area/nucleus and a 3.1-fold decrease of satellite cells (SCs) occurred. We propose that during the pre-competitive season SC proliferation occurred following with increased cell fusion during the competitive season. Expression of twenty-two envelope genes of muscle biopsies demonstrated a significant increase of putative muscle-cell fusogenic genes Syncytin-1 and Syncytin-3, but also for the non-fusogenic erv3. Immunohistochemistry analyses showed that Syncytin-1 mainly localized to the sarcolemma of myofibers positive for myosin heavy-chain isotypes. Cellular receptors SLC1A4 and SLC1A5 of Syncytin-1 showed significant decrease of expression in post-competitive muscles compared with the pre-competitive season, but only SLC1A4 protein expression localized throughout the myofiber. Erv3 protein was strongly expressed throughout the myofiber, whereas envK1-7 localized to SC nuclei and myonuclei. Syncytin-1 transcription factors, PPARγ and RXRα, showed no protein expression in the myofiber, whereas the pCREB-Ser133 activator of Syncytin-1 was enriched to SC nuclei and myonuclei. Syncytin-1, Syncytin-3, SLC1A4 and PAX7 gene regulations along with MyoD1 and myogenin were verified during proliferating or actively-fusing human primary myoblast cell cultures, resembling muscle biopsies of cyclists. Myoblast treatment with anti-Synycytin-1 abrogated cell fusion in vitro. Our findings support functional roles for ERV envelope proteins, especially Syncytin-1, contributing to cell fusion of myotubes.
Collapse
Affiliation(s)
- Sebastian Frese
- Institute of Cardiovascular Research and Sport Medicine, Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Am Sportpark Muengersdorf, Cologne, Germany
- University Hospital Zurich, Department of Neurology, Frauenklinikstrasse, Zurich, Switzerland
- Institute of Human Movement Sciences and Sport, Exercise Physiology, ETH Zurich, Winterthurerstrasse, Zurich, Switzerland
| | - Matthias Ruebner
- Friedrich-Alexander University Erlangen-Nürnberg, University-Clinic Erlangen, Department of Gynaecology and Obstetrics, Laboratory for Molecular Medicine, Erlangen, Universitaetsstrasse, Erlangen, Germany
| | - Frank Suhr
- Institute of Cardiovascular Research and Sport Medicine, Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Am Sportpark Muengersdorf, Cologne, Germany
| | - Thierry M. Konou
- Institute of Cardiovascular Research and Sport Medicine, Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Am Sportpark Muengersdorf, Cologne, Germany
| | - Kim A. Tappe
- Institute of Cardiovascular Research and Sport Medicine, Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Am Sportpark Muengersdorf, Cologne, Germany
| | - Marco Toigo
- Institute of Human Movement Sciences and Sport, Exercise Physiology, ETH Zurich, Winterthurerstrasse, Zurich, Switzerland
- University of Zurich, Balgrist University Hospital, Department of Orthopaedics, Forchstrasse, Zurich, Switzerland
| | - Hans H. Jung
- University Hospital Zurich, Department of Neurology, Frauenklinikstrasse, Zurich, Switzerland
| | - Christine Henke
- Friedrich-Alexander University Erlangen-Nürnberg, University-Clinic Erlangen, Department of Gynaecology and Obstetrics, Laboratory for Molecular Medicine, Erlangen, Universitaetsstrasse, Erlangen, Germany
| | - Ruth Steigleder
- Friedrich-Alexander University Erlangen-Nürnberg, University-Clinic Erlangen, Department of Gynaecology and Obstetrics, Laboratory for Molecular Medicine, Erlangen, Universitaetsstrasse, Erlangen, Germany
| | - Pamela L. Strissel
- Friedrich-Alexander University Erlangen-Nürnberg, University-Clinic Erlangen, Department of Gynaecology and Obstetrics, Laboratory for Molecular Medicine, Erlangen, Universitaetsstrasse, Erlangen, Germany
| | - Hanna Huebner
- Friedrich-Alexander University Erlangen-Nürnberg, University-Clinic Erlangen, Department of Gynaecology and Obstetrics, Laboratory for Molecular Medicine, Erlangen, Universitaetsstrasse, Erlangen, Germany
| | - Matthias W. Beckmann
- Friedrich-Alexander University Erlangen-Nürnberg, University-Clinic Erlangen, Department of Gynaecology and Obstetrics, Laboratory for Molecular Medicine, Erlangen, Universitaetsstrasse, Erlangen, Germany
| | - Piet van der Keylen
- Institute of Anatomy, Friedrich-Alexander University of Erlangen-Nürnberg, Krankenhausstrasse, Erlangen, Germany
| | - Benedikt Schoser
- Ludwig Maximilian University Munich, Department of Neurology, Friedrich Baur Institute, Ziemssenstrasse, Munich, Germany
| | - Thorsten Schiffer
- German Sport University Cologne, Outpatient Clinic for Sports Traumatology and Public Health Consultation, Am Sportpark Muengersdorf, Cologne, Germany
| | - Laura Frese
- University Hospital and University Zurich, Division of Surgical Research, Raemistrasse, Zurich, Switzerland
| | - Wilhelm Bloch
- Institute of Cardiovascular Research and Sport Medicine, Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Am Sportpark Muengersdorf, Cologne, Germany
- The German Research Centre of Elite Sport, German Sport University Cologne, Am Sportpark Muengersdorf, Cologne, Germany
| | - Reiner Strick
- Friedrich-Alexander University Erlangen-Nürnberg, University-Clinic Erlangen, Department of Gynaecology and Obstetrics, Laboratory for Molecular Medicine, Erlangen, Universitaetsstrasse, Erlangen, Germany
- * E-mail:
| |
Collapse
|
38
|
Stiles TL, Kapiloff MS, Goldberg JL. The role of soluble adenylyl cyclase in neurite outgrowth. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1842:2561-8. [PMID: 25064589 PMCID: PMC4262618 DOI: 10.1016/j.bbadis.2014.07.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 07/11/2014] [Accepted: 07/15/2014] [Indexed: 12/25/2022]
Abstract
Axon regeneration in the mature central nervous system is limited by extrinsic inhibitory signals and a postnatal decline in neurons' intrinsic growth capacity. Neuronal levels of the second messenger cAMP are important in regulating both intrinsic growth capacity and neurons' responses to extrinsic factors. Approaches which increase intracellular cAMP in neurons enhance neurite outgrowth and facilitate regeneration after injury. Thus, understanding the factors which affect cAMP in neurons is of potential therapeutic importance. Recently, soluble adenylyl cyclase (sAC, ADCY10), the ubiquitous, non-transmembrane adenylyl cyclase, was found to play a key role in neuronal survival and axon growth. sAC is activated by bicarbonate and cations and may translate physiologic signals from metabolism and electrical activity into a neuron's decision to survive or regenerate. Here we critically review the literature surrounding sAC and cAMP signaling in neurons to further elucidate the potential role of sAC signaling in neurite outgrowth and regeneration. This article is part of a Special Issue entitled: The role of soluble adenylyl cyclase in health and disease.
Collapse
Affiliation(s)
- Travis L Stiles
- Shiley Eye Center, University of California, San Diego, CA 92093, USA
| | - Michael S Kapiloff
- Departments of Pediatrics and Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | | |
Collapse
|
39
|
Tanabe K, Matsushima-Nishiwaki R, Kozawa O, Iida H. Dexmedetomidine suppresses interleukin-1β-induced interleukin-6 synthesis in rat glial cells. Int J Mol Med 2014; 34:1032-8. [PMID: 25069417 DOI: 10.3892/ijmm.2014.1863] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 07/18/2014] [Indexed: 11/06/2022] Open
Abstract
Dexmedetomidine, an α2-adrenoceptor agonist, is used as a sedative medication for criticalyl ill patients and is known to exert neuroprotective effects by direct action on neurons and indirect action on neurons through astrocytes. Interleukin (IL)-6 plays a key role in neuroinflammation, which accompanies infection, traumatic brain injury, ischemia, neurodegenerative disorders, as both a pro-inflammatory cytokine and an anti-inflammatory cytokine. Dexmedetomidine suppresses immune function. However, the effects of dexmedetomidine on cytokine synthesis in the central nervous system (CNS) remain elusive. We previously reported that IL-1β stimulates IL-6 synthesis in the rat C6 glioma cell line through the phosphorylation of p38 mitogen-activated protein (MAP) kinase, stress-activated protein kinase (SAPK)/c-Jun N-terminal kinase (JNK) and IκB. In the present study, we investigated the effects of dexmedetomidine on the IL-1β-induced IL-6 synthesis in C6 cells. Dexmedetomidine inhibited the IL-1β-stimulated IL-6 release and mRNA expression in C6 cells. 8-Bromo-adenosine-3',5'-cyclic monophosphate, but not 8-bromo-guanosine 3',5'-cyclic monophosphate, significantly enhanced the IL-1β-induced IL-6 release and mRNA expression. However, dexmedetomidine failed to affect cAMP accumulation in the cells treated with IL-1β or forskolin, an activator of adenylyl cyclase. Yohimbine, an α2-adrenoceptor antagonist, did not reverse the suppressive effects of dexmedetomidine on the IL-1β-induced IL-6 release. Dexmedetomidine did not affect the IL-1β-induced phosphorylation of p38 MAP kinase, SAPK/JNK, IκB, nuclear factor (NF)-κB or c-Jun. Our findings strongly suggest that dexmedetomidine inhibits the IL-1β-induced IL-6 synthesis independently of the adenylyl cyclase-cAMP pathway through α2-adrenoceptors in C6 glioma cells. It is possible that dexmedetomidine may affect the immune system in the CNS by regulating the production of IL-6.
Collapse
Affiliation(s)
- Kumiko Tanabe
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | | | - Osamu Kozawa
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Hiroki Iida
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| |
Collapse
|
40
|
Wang Q, Fong R, Mason P, Fox AP, Xie Z. Caffeine accelerates recovery from general anesthesia. J Neurophysiol 2014; 111:1331-40. [PMID: 24375022 PMCID: PMC3949308 DOI: 10.1152/jn.00792.2013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 12/26/2013] [Indexed: 11/22/2022] Open
Abstract
General anesthetics inhibit neurotransmitter release from both neurons and secretory cells. If inhibition of neurotransmitter release is part of an anesthetic mechanism of action, then drugs that facilitate neurotransmitter release may aid in reversing general anesthesia. Drugs that elevate intracellular cAMP levels are known to facilitate neurotransmitter release. Three cAMP elevating drugs (forskolin, theophylline, and caffeine) were tested; all three drugs reversed the inhibition of neurotransmitter release produced by isoflurane in PC12 cells in vitro. The drugs were tested in isoflurane-anesthetized rats. Animals were injected with either saline or saline containing drug. All three drugs dramatically accelerated recovery from isoflurane anesthesia, but caffeine was most effective. None of the drugs, at the concentrations tested, had significant effects on breathing rates, O2 saturation, heart rate, or blood pressure in anesthetized animals. Caffeine alone was tested on propofol-anesthetized rats where it dramatically accelerated recovery from anesthesia. The ability of caffeine to accelerate recovery from anesthesia for different chemical classes of anesthetics, isoflurane and propofol, opens the possibility that it will do so for all commonly used general anesthetics, although additional studies will be required to determine whether this is in fact the case. Because anesthesia in rodents is thought to be similar to that in humans, these results suggest that caffeine might allow for rapid and uniform emergence from general anesthesia in human patients.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Neurobiology, Pharmacology and Physiology, University of Chicago, Chicago, Illinois
| | | | | | | | | |
Collapse
|
41
|
Chen M, Zhang X, Xu H, Ma X, Jiang W, Xu T. Inhibitory effect of spinal mGlu(5) receptor antisense oligonucleotide on the up-regulated expression of spinal G protein associated with chronic morphine treatment. Eur J Pharmacol 2013; 723:253-8. [PMID: 24296320 DOI: 10.1016/j.ejphar.2013.11.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 11/12/2013] [Accepted: 11/20/2013] [Indexed: 11/19/2022]
Abstract
Knockdown of spinal metabotropic glutamate 5 (mGlu5) receptor was shown to inhibit the development of intrathecal morphine antinociceptive tolerance. The present work was designed to evaluate the expression of spinal G-protein during morphine tolerance and knockdown of spinal mGlu5 receptor with antisense oligonucleotide (ODN). Rats were treated with saline, morphine, mGlu5 receptor antisense or mismatch ODN intrathecally. Behavioral tests were employed to test the thermal and mechanical pain thresholds. Five days later, rats were scarified and spinal expression of spinal Gαi, Gαo, Gαq and Gβ were detected. Consistent with the previous results, knockdown of spinal mGlu5 receptor could inhibit spinal morphine antinociceptive tolerance in behavioral tests (P<0.05). The mGlu5 receptor antisense ODN produced a significant reduction in mGlu5 receptor protein of about 56.6% compared with the control group (P<0.05). Expression of spinal Gαi, Gαo, Gαq and Gβ were up-regulated while morphine tolerance developed (P<0.05). Antisense ODN of spinal mGlu5 receptor, but not mismatched ODN, reduced the spinal dorsal horn levels of Gαi, Gαo, Gαs, Gαq and Gβ (P<0.05). We conclude that expression of spinal G (αi, αo, αs, αq and β) protein may be up-regulated after chronic morphine treatment which could be attenuated by knockdown of spinal mGlu5 receptor with antisense ODN.
Collapse
Affiliation(s)
- Moxi Chen
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 600 Yi Shan Road, Shanghai 200233, China
| | - Xiaoli Zhang
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 600 Yi Shan Road, Shanghai 200233, China
| | - Hao Xu
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 600 Yi Shan Road, Shanghai 200233, China
| | - Xiaqing Ma
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 600 Yi Shan Road, Shanghai 200233, China
| | - Wei Jiang
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 600 Yi Shan Road, Shanghai 200233, China.
| | - Tao Xu
- Department of Anesthesiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University, 600 Yi Shan Road, Shanghai 200233, China.
| |
Collapse
|
42
|
Dlouhá K, Kagan D, Roubalová L, Ujčíková H, Svoboda P. Plasma membrane density of GABA(B)-R1a, GABA(B)-R1b, GABA-R2 and trimeric G-proteins in the course of postnatal development of rat brain cortex. Physiol Res 2013; 62:547-59. [PMID: 24020808 DOI: 10.33549/physiolres.932552] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
With the aim to understand the onset of expression and developmental profile of plasma membrane (PM) content /density of crucial components of GABA(B)-R signaling cascade, GABA(B)-R1a, GABA(B)-R1b, GABA(B)-R2, G(i)1/G(i)2alpha, G(i)3alpha, G(o)alpha, G(z)alpha and Gbeta subunit proteins were determined by quantitative immunoblotting and compared in PM isolated from brain cortex of rats of different ages: between postnatal-day-1 (PD1) and 90 (PD90). PM density of GABA(B)-R1a, GABA(B)-R2, G(i)1/G(i)2alpha, G(i)3alpha, G(o)alpha, G(z)alpha and Gbeta was high already at birth and further development was reflected in parallel decrease of both GABA(B)-R1a and GABA(B)-R2 subunits. The major decrease of GABA(B)-R1a and GABA(B)-R2 occurred between the birth and PD15: to 55 % (R1a, **) and 51 % (R2, **), respectively. Contrarily, PM level of the cognate G-proteins G(i)1/G(i)2alpha, G(i)3alpha, G(o)alpha, G(z)alpha and Gbeta was unchanged in the course of the whole postnatal period of brain cortex development. Maturation of GABA(B)-R cascade was substantially different from ontogenetic profile of prototypical plasma membrane marker, Na, K-ATPase, which was low at birth and further development was reflected in continuous increase of PM density of this enzyme. Major change occurred between the birth and PD25. In adult rats, membrane content of Na, K-ATPase was 3-times higher than around the birth.
Collapse
Affiliation(s)
- K Dlouhá
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic.
| | | | | | | | | |
Collapse
|
43
|
Gassmann M, Bettler B. Regulation of neuronal GABA(B) receptor functions by subunit composition. Nat Rev Neurosci 2012; 13:380-94. [PMID: 22595784 DOI: 10.1038/nrn3249] [Citation(s) in RCA: 259] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
GABA(B) receptors (GABA(B)Rs) are G protein-coupled receptors for GABA, the main inhibitory neurotransmitter in the CNS. In the past 5 years, notable advances have been made in our understanding of the molecular composition of these receptors. GABA(B)Rs are now known to comprise principal and auxiliary subunits that influence receptor properties in distinct ways. The principal subunits regulate the surface expression and the axonal versus dendritic distribution of these receptors, whereas the auxiliary subunits determine agonist potency and the kinetics of the receptor response. This Review summarizes current knowledge on how the subunit composition of GABA(B)Rs affects the distribution of these receptors, neuronal processes and higher brain functions.
Collapse
Affiliation(s)
- Martin Gassmann
- Department of Biomedicine, Institute of Physiology, University of Basel, Klingelbergstr. 50-70, 4056 Basel, Switzerland.
| | | |
Collapse
|
44
|
Axelband F, Dias J, Miranda F, Ferrão FM, Reis RI, Costa-Neto CM, Lara LS, Vieyra A. Angiotensin-(3-4) counteracts the Angiotensin II inhibitory action on renal Ca2+-ATPase through a cAMP/PKA pathway. ACTA ACUST UNITED AC 2012; 177:27-34. [PMID: 22561691 DOI: 10.1016/j.regpep.2012.04.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Revised: 03/30/2012] [Accepted: 04/23/2012] [Indexed: 12/14/2022]
Abstract
We recently demonstrated that Angiotensin-(3-4) [Ang-(3-4)], an Ang II-derived dipeptide, overcomes inhibition of plasma membrane Ca(2+)-ATPase promoted by nanomolar concentrations of Ang II in basolateral membranes of renal proximal tubule cells, with involvement of a so far unknown AT(2)R-dependent and NO-independent mechanism. The present study investigates the signaling pathway triggered by Ang-(3-4) that is responsible for counteracting the inhibitory effect of Ang II, and attempts to elucidate the functional interaction of the dipeptide with Ang II at the level of AT(2)R. Stimulation by cholera toxin of G(s)α protein structurally linked to AT(2)R--as revealed by their co-immunoprecipitation--mimicked the effect of Ang-(3-4) on Ca(2+)-ATPase activity. Furthermore, addition of dibutyril-cAMP (db-cAMP) mimicked Ang-(3-4), whereas the specific PKA inhibitor, PKAi(5-24) peptide, suppressed the counter-regulatory effect of Ang-(3-4) and the AT(2)R agonist, CGP42112A. Membrane-associated PKA activity was stimulated by Ang-(3-4) or CGP42112A to comparable levels as db-cAMP, and the Ang-(3-4) effect was abrogated by the AT(2)R antagonist PD123319, whereas the AT(1)R antagonist Losartan had no effect. Ang-(3-4) stimulated PKA-mediated phosphorylation of Ca(2+)-ATPase and activated PKA to comparable levels. Binding assays demonstrated that Ang-(3-4) could not displace (3)H-Ang II from HEK 293T cells expressing AT(2)R, but 10(-10) mol/L Ang-(3-4) resulted in the appearance of a probable higher-affinity site (picomolar range) for Ang II. The results presented herein demonstrate that Ang-(3-4), acting as an allosteric enhancer, suppresses Ang II-mediated inhibition of Ca(2+)-ATPase through an AT(2)R/cAMP/PKA pathway, after inducing conformational changes in AT(2)R that results in generation of higher-affinity sites for Ang II.
Collapse
Affiliation(s)
- Flavia Axelband
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Lanzafame AA, Christopoulos A, Mitchelson F. Cellular Signaling Mechanisms for Muscarinic Acetylcholine Receptors. ACTA ACUST UNITED AC 2011. [DOI: 10.3109/10606820308263] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
46
|
Lee CWS, Yan JY, Chiang YC, Hung TW, Wang HL, Chiou LC, Ho IK. Differential pharmacological actions of methadone and buprenorphine in human embryonic kidney 293 cells coexpressing human μ-opioid and opioid receptor-like 1 receptors. Neurochem Res 2011; 36:2008-21. [PMID: 21671107 PMCID: PMC3183316 DOI: 10.1007/s11064-011-0525-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2011] [Indexed: 11/26/2022]
Abstract
Methadone and buprenorphine are used in maintenance therapy for heroin addicts. In this study, we compared their effects on adenylate cyclase (AC) activity in human embryonic kidney (HEK) 293 cells stably overexpressing human μ-opioid receptor (MOR) and nociceptin/opioid receptor-like 1 receptor (ORL1) simultaneously. After acute exposure, methadone inhibited AC activity; however, buprenorphine induced compromised AC inhibition. When naloxone was introduced after 30 min incubation with methadone, the AC activity was enhanced. This was not observed in the case of buprenorphine. Enhancement of the AC activity was more significant when the incubation lasted for 4 h, and prolonged exposure to buprenorphine elevated the AC activity as well. The removal of methadone and buprenorphine by washing also obtained similar AC superactivation as that revealed by naloxone challenge. The study demonstrated that methadone and buprenorphine exert initially different yet eventually convergent adaptive changes of AC activity in cells coexpressing human MOR and ORL1 receptors.
Collapse
Affiliation(s)
- Cynthia Wei-Sheng Lee
- Division of Mental Health and Addiction Medicine, Institute of Population Health Sciences, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County, 35053 Taiwan
| | - Jia-Ying Yan
- Division of Mental Health and Addiction Medicine, Institute of Population Health Sciences, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County, 35053 Taiwan
| | - Yao-Chang Chiang
- Division of Mental Health and Addiction Medicine, Institute of Population Health Sciences, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County, 35053 Taiwan
| | - Tsai-Wei Hung
- Division of Mental Health and Addiction Medicine, Institute of Population Health Sciences, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County, 35053 Taiwan
| | - Hung-Li Wang
- Department of Physiology, Chang Gung University School of Medicine, Kwei-San, Taoyuan, 33302 Taiwan
| | - Lih-Chu Chiou
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051 Taiwan
| | - Ing-Kang Ho
- Division of Mental Health and Addiction Medicine, Institute of Population Health Sciences, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County, 35053 Taiwan
| |
Collapse
|
47
|
Kimple AJ, Bosch DE, Giguère PM, Siderovski DP. Regulators of G-protein signaling and their Gα substrates: promises and challenges in their use as drug discovery targets. Pharmacol Rev 2011; 63:728-49. [PMID: 21737532 PMCID: PMC3141876 DOI: 10.1124/pr.110.003038] [Citation(s) in RCA: 190] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Because G-protein coupled receptors (GPCRs) continue to represent excellent targets for the discovery and development of small-molecule therapeutics, it is posited that additional protein components of the signal transduction pathways emanating from activated GPCRs themselves are attractive as drug discovery targets. This review considers the drug discovery potential of two such components: members of the "regulators of G-protein signaling" (RGS protein) superfamily, as well as their substrates, the heterotrimeric G-protein α subunits. Highlighted are recent advances, stemming from mouse knockout studies and the use of "RGS-insensitivity" and fast-hydrolysis mutations to Gα, in our understanding of how RGS proteins selectively act in (patho)physiologic conditions controlled by GPCR signaling and how they act on the nucleotide cycling of heterotrimeric G-proteins in shaping the kinetics and sensitivity of GPCR signaling. Progress is documented regarding recent activities along the path to devising screening assays and chemical probes for the RGS protein target, not only in pursuits of inhibitors of RGS domain-mediated acceleration of Gα GTP hydrolysis but also to embrace the potential of finding allosteric activators of this RGS protein action. The review concludes in considering the Gα subunit itself as a drug target, as brought to focus by recent reports of activating mutations to GNAQ and GNA11 in ocular (uveal) melanoma. We consider the likelihood of several strategies for antagonizing the function of these oncogene alleles and their gene products, including the use of RGS proteins with Gα(q) selectivity.
Collapse
Affiliation(s)
- Adam J Kimple
- Department of Pharmacology, UNC Neuroscience Center, UNC School of Medicine, University of North Carolina at Chapel Hill, 120 Mason Farm Road, Suite 4010, Chapel Hill, NC 27599-7365, USA
| | | | | | | |
Collapse
|
48
|
Wang Y, Kong Y, Shei GJ, Kang L, Cvijic ME. Development of a cyclic adenosine monophosphate assay for Gi-coupled G protein-coupled receptors by utilizing the endogenous calcitonin activity in Chinese hamster ovary cells. Assay Drug Dev Technol 2011; 9:522-31. [PMID: 21561374 DOI: 10.1089/adt.2010.0361] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Activation of G(i)-coupled G protein-coupled receptor (GPCRs) by their ligands leads to inhibition of adenylyl cyclase (AC) and reduction of cyclic adenosine monophosphate (cAMP) levels in cells. The traditional cAMP assay for G(i)-coupled GPCRs commonly uses forskolin, a nonspecific AC activator, to increase the basal cAMP level in cells to create an assay window for ligand detection. However, there is still a need to develop a nonforskolin-based cAMP assay because of the challenges inherent in titrating the concentration of forskolin to achieve a reliable assay window, along with issues related to the cAMP-independent effects of forskolin. Herein, we describe such an assay by utilizing the endogenous activity of the calcitonin receptor in Chinese hamster ovary (CHO) cells. The calcitonin receptor is a G(s)-coupled GPCR that, when activated by calcitonin, leads to the stimulation of AC and increases cAMP in cells. Thus, we use calcitonin, instead of forskolin, to increase the basal cAMP level in CHO cells to achieve an assay window. We demonstrated that calcitonin peptides robustly increased cAMP accumulation in several CHO cell lines stably expressing well-known G(i)-coupled GPCRs, such as the Dopamine D2 receptor, the Opioid μ receptor, or the Cannabinoid receptor-1. Agonists of these G(i)-coupled GPCRs attenuated calcitonin-induced cAMP production in their receptor stable cell lines. On the other hand, antagonists and/or inverse agonists blocked the effects of their agonists on calcitonin-induced cAMP production. This calcitonin-based cAMP assay has been demonstrated to be sensitive and robust and exhibited acceptable assay windows (signal/noise ratio) and, thus, can be applied to screen for agonists and antagonists/inverse agonists of G(i)-coupled GPCRs in high-throughput screening formats.
Collapse
Affiliation(s)
- Yuren Wang
- Metabolic Disease Research, Bristol-Myers Squibb Company, 311 Pennington-Rocky Hill Road, Pennington, NJ 08534, USA.
| | | | | | | | | |
Collapse
|
49
|
Zalduegui A, López de Jesús M, Callado LF, Meana JJ, Sallés J. Levels of Gsα(short and long), Gα(olf) and Gβ(common) subunits, and calcium-sensitive adenylyl cyclase isoforms (1, 5/6, 8) in post-mortem human brain caudate and cortical membranes: comparison with rat brain membranes and potential stoichiometric relationships. Neurochem Int 2010; 58:180-9. [PMID: 21115086 DOI: 10.1016/j.neuint.2010.11.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Revised: 11/12/2010] [Accepted: 11/21/2010] [Indexed: 11/24/2022]
Abstract
The levels of expression of Gsα(short and long), Gα(olf) and Gβ(common) subunits, and calcium-sensitive adenylyl cyclases isoforms (AC1, 5/6, and 8) in human brain cortical and caudate membranes were quantified by western blot analysis in order to establish their contribution to the patterns of AC functioning. Both areas expressed Gsα(long) (52 kDa) with values ranging from about 1400 ng/mg of membrane protein in cerebral cortex to close to 600 ng/mg of membrane protein in caudate nucleus. In contrast, Gsα(short) and Gsα(olf) were expressed separately, Gsα(short) in cortical membranes with values around 500 ng/mg of membrane protein and Gα(olf) in caudate membranes with values around 1300 ng/mg of membrane protein. Quantitative measurements of Gβ, revealed a similar expression level in cortical and caudate membranes (5444±732 versus 5511±394 ng/mg protein; p=0.966). The B(max) values of GTPγS-dependent [(3)H]-forskolin binding show the following descending order: rat striatal membranes>rat cortical membranes=human caudate membranes>human cortical membranes. Therefore, as measured immunochemically and by [(3)H]-forskolin binding, there seems to be a vast excess of Gsα subunits over catalytic units of AC. The highest levels of AC5/6 expression were detected in caudate membranes. AC8 was little expressed, and there were no significant differences in the relative values between both human brain regions. Finally, the levels of the AC1 isoform were significantly lower in caudate than in cortical membranes. It is concluded that these stoichiometric data contribute nonetheless to explain the significant differences observed in signalling capacities through the AC system in both human brain regions.
Collapse
Affiliation(s)
- Amaia Zalduegui
- Department of Pharmacology, Faculty of Pharmacy (Vitoria-Gasteiz), University of the Basque Country (UPV/EHU), Spain
| | | | | | | | | |
Collapse
|
50
|
Ho D, Yan L, Iwatsubo K, Vatner DE, Vatner SF. Modulation of beta-adrenergic receptor signaling in heart failure and longevity: targeting adenylyl cyclase type 5. Heart Fail Rev 2010; 15:495-512. [PMID: 20658186 PMCID: PMC3655553 DOI: 10.1007/s10741-010-9183-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Despite remarkable advances in therapy, heart failure remains a leading cause of morbidity and mortality. Although enhanced beta-adrenergic receptor stimulation is part of normal physiologic adaptation to either the increase in physiologic demand or decrease in cardiac function, chronic beta-adrenergic stimulation has been associated with increased mortality and morbidity in both animal models and humans. For example, overexpression of cardiac Gsalpha or beta-adrenergic receptors in transgenic mice results in enhanced cardiac function in young animals, but with prolonged overstimulation of this pathway, cardiomyopathy develops in these mice as they age. Similarly, chronic sympathomimetic amine therapy increases morbidity and mortality in patients with heart failure. Conversely, the use of beta-blockade has proven to be of benefit and is currently part of the standard of care for heart failure. It is conceivable that interrupting distal mechanisms in the beta-adrenergic receptor-G protein-adenylyl cyclase pathway may also provide targets for future therapeutic modalities for heart failure. Interestingly, there are two major isoforms of adenylyl cyclase (AC) in the heart (type 5 and type 6), which may exert opposite effects on the heart, i.e., cardiac overexpression of AC6 appears to be protective, whereas disruption of type 5 AC prolongs longevity and protects against cardiac stress. The goal of this review is to summarize the paradigm shift in the treatment of heart failure over the past 50 years from administering sympathomimetic amine agonists to administering beta-adrenergic receptor antagonists, and to explore the basis for a novel therapy of inhibiting type 5 AC.
Collapse
Affiliation(s)
- David Ho
- Department of Cell Biology and Molecular Medicine and The Cardiovascular Research Institute, University of Medicine & Dentistry of New Jersey, New Jersey Medical School, 185 South Orange Avenue, MSB G609, Newark, NJ 07103, USA
| | | | | | | | | |
Collapse
|