1
|
Martins IRR, de Melo Medeiros M, Vasconcelos LHC, Silva MDCC, Queiroga FR, Cavalcante-Silva LHA, da Costa Nunes IK, Lima LM, da Silva PM, Silva BAD. New phosphodiesterase-4 inhibitors present airways relaxant activity in a guinea pig acute asthma model. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:4419-4434. [PMID: 38108836 DOI: 10.1007/s00210-023-02905-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
Asthma is a disease characterized by chronic inflammation and hyper responsiveness of airways. We aimed to assess the relaxant potential of phosphodiesterase-4 (PDE4) inhibitors N-sulfonilhidrazonic derivatives on non-asthmatic and asthmatic guinea pig trachea. Firstly, guinea pigs were sensitized and challenged with ovalbumin, and then morphological, and contractile changes were evaluated resulting from asthma, followed by evaluation of relaxant effect of derivatives on guinea pig trachea and the cAMP levels measurement by ELISA. It has been evidenced hypertrophy of airway smooth muscle, inflammatory infiltrate, and vascular abnormalities. Moreover, only sensitized tracheal rings were responsive to OVA. Contractile response to histamine, but not to carbachol, was greater in sensitized animals, however the relaxant response to aminophylline and isoprenaline were the same in non-asthmatics and asthmatics. N-sulfonilhidrazonic derivatives presented equipotent relaxant action independent of epithelium, with exception of LASSBio-1850 that presented a low efficacy (< 50%) and LASSBio-1847 with a 4-fold higher potency on asthmatics. LASSBio-1847 relaxant curve was impaired in the presence of propranolol and potentiated by isoprenaline in both groups. Furthermore, relaxation was potentiated 54- and 4-fold by forskolin in non-asthmatics and asthmatics, respectively. Likewise, LASSBio-1847 potentiated relaxant curve of aminophylline 147- and 4-fold in both groups. The PKA inhibitor H-89 impaired the relaxant potency of the derivative. Finally, LASSBio-1847 increased tracheal intracellular cAMP levels similarly to rolipram, selective PDE4 inhibitor, in both animals. LASSBio-1847 showed to be promising to relax guinea pig trachea from non-sensitized and sensitized guinea pigs by activation of β2-adrenergic receptors/AC/cAMP pathway.
Collapse
Affiliation(s)
- Italo Rossi Roseno Martins
- Laboratório de Farmacologia Funcional Professor George Thomas, Instituto de Pesquisa em Fármacos e Medicamentos, Universidade Federal da Paraíba, João Pessoa, Brazil.
- Departamento de Medicina, Campus Senador Helvídio Nunes de Barros, Universidade Federal do Piauí, Picos, PI, 64607-670, Brazil.
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Brazil.
| | - Millena de Melo Medeiros
- Laboratório de Farmacologia Funcional Professor George Thomas, Instituto de Pesquisa em Fármacos e Medicamentos, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Luiz Henrique César Vasconcelos
- Laboratório de Farmacologia Funcional Professor George Thomas, Instituto de Pesquisa em Fármacos e Medicamentos, Universidade Federal da Paraíba, João Pessoa, Brazil
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Brazil
- Departamento de Fisiologia e Patologia, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Maria da Conceição Correia Silva
- Laboratório de Farmacologia Funcional Professor George Thomas, Instituto de Pesquisa em Fármacos e Medicamentos, Universidade Federal da Paraíba, João Pessoa, Brazil
- PostDoc Conselho Nacional de Desenvolvimento Científico e Tecnológico/Fundação de Amparo a Ciência e Tecnologia de Pernambuco, Recife, Brazil
| | - Fernando Ramos Queiroga
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Brazil
- Laboratório de Imunologia e Patologia de Invertebrados, Departamento de Biologia Molecular, Centro de Ciências Exatas e da Natureza, Universidade Federal da Paraíba, João Pessoa, Brazil
| | | | - Isabelle Karine da Costa Nunes
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio ®), Instituto Nacional de Ciência e Tecnologia de Fármacos e Medicamentos (INCT-INOFAR), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lidia Moreira Lima
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio ®), Instituto Nacional de Ciência e Tecnologia de Fármacos e Medicamentos (INCT-INOFAR), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia Mirella da Silva
- Laboratório de Imunologia e Patologia de Invertebrados, Departamento de Biologia Molecular, Centro de Ciências Exatas e da Natureza, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Bagnólia Araújo da Silva
- Laboratório de Farmacologia Funcional Professor George Thomas, Instituto de Pesquisa em Fármacos e Medicamentos, Universidade Federal da Paraíba, João Pessoa, Brazil
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Brazil
- Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Brazil
| |
Collapse
|
2
|
Bock A, Irannejad R, Scott JD. cAMP signaling: a remarkably regional affair. Trends Biochem Sci 2024; 49:305-317. [PMID: 38310024 PMCID: PMC11175624 DOI: 10.1016/j.tibs.2024.01.004] [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: 11/02/2023] [Revised: 12/22/2023] [Accepted: 01/10/2024] [Indexed: 02/05/2024]
Abstract
Louis Pasteur once famously said 'in the fields of observation chance favors only the prepared mind'. Much of chance is being in the right place at the right time. This is particularly true in the crowded molecular environment of the cell where being in the right place is often more important than timing. Although Brownian motion argues that enzymes will eventually bump into substrates, this probability is greatly enhanced if both molecules reside in the same subcellular compartment. However, activation of cell signaling enzymes often requires the transmission of chemical signals from extracellular stimuli to intracellular sites of action. This review highlights new developments in our understanding of cAMP generation and the 3D utilization of this second messenger inside cells.
Collapse
Affiliation(s)
- Andreas Bock
- Rudolf Boehm Institute of Pharmacology and Toxicology, Medical Faculty, Leipzig University, 04107 Leipzig, Germany.
| | - Roshanak Irannejad
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - John D Scott
- Department of Pharmacology, University of Washington Medical Center, Seattle, WA 98195, USA.
| |
Collapse
|
3
|
Johnson SC, Annamdevula NS, Leavesley SJ, Francis CM, Rich TC. Hyperspectral imaging and dynamic region of interest tracking approaches to quantify localized cAMP signals. Biochem Soc Trans 2024; 52:191-203. [PMID: 38334148 PMCID: PMC11115359 DOI: 10.1042/bst20230352] [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: 11/15/2023] [Revised: 01/10/2024] [Accepted: 01/15/2024] [Indexed: 02/10/2024]
Abstract
Cyclic adenosine monophosphate (cAMP) is a ubiquitous second messenger known to orchestrate a myriad of cellular functions over a wide range of timescales. In the last 20 years, a variety of single-cell sensors have been developed to measure second messenger signals including cAMP, Ca2+, and the balance of kinase and phosphatase activities. These sensors utilize changes in fluorescence emission of an individual fluorophore or Förster resonance energy transfer (FRET) to detect changes in second messenger concentration. cAMP and kinase activity reporter probes have provided powerful tools for the study of localized signals. Studies relying on these and related probes have the potential to further revolutionize our understanding of G protein-coupled receptor signaling systems. Unfortunately, investigators have not been able to take full advantage of the potential of these probes due to the limited signal-to-noise ratio of the probes and the limited ability of standard epifluorescence and confocal microscope systems to simultaneously measure the distributions of multiple signals (e.g. cAMP, Ca2+, and changes in kinase activities) in real time. In this review, we focus on recently implemented strategies to overcome these limitations: hyperspectral imaging and adaptive thresholding approaches to track dynamic regions of interest (ROI). This combination of approaches increases signal-to-noise ratio and contrast, and allows identification of localized signals throughout cells. These in turn lead to the identification and quantification of intracellular signals with higher effective resolution. Hyperspectral imaging and dynamic ROI tracking approaches offer investigators additional tools with which to visualize and quantify multiplexed intracellular signaling systems.
Collapse
Affiliation(s)
- Santina C Johnson
- Department of Pharmacology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, U.S.A
- Center for Lung Biology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, U.S.A
| | - Naga S Annamdevula
- Department of Pharmacology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, U.S.A
- Department of Physiology and Cell Biology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, U.S.A
- Center for Lung Biology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, U.S.A
| | - Silas J Leavesley
- Department of Pharmacology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, U.S.A
- Center for Lung Biology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, U.S.A
- Chemical and Biomolecular Engineering, University of South Alabama, Mobile, AL, U.S.A
| | - C Michael Francis
- Department of Physiology and Cell Biology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, U.S.A
- Center for Lung Biology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, U.S.A
| | - Thomas C Rich
- Department of Pharmacology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, U.S.A
- Center for Lung Biology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, U.S.A
| |
Collapse
|
4
|
Cattani-Cavalieri I, Li Y, Margolis J, Bogard A, Roosan MR, Ostrom RS. Quantitative phosphoproteomic analysis reveals unique cAMP signaling pools emanating from AC2 and AC6 in human airway smooth muscle cells. Front Physiol 2023; 14:1149063. [PMID: 36926196 PMCID: PMC10011497 DOI: 10.3389/fphys.2023.1149063] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 02/16/2023] [Indexed: 03/08/2023] Open
Abstract
Human airway smooth muscle (HASM) is the primary target of ßAR agonists used to control airway hypercontractility in asthma and chronic obstructive pulmonary disease (COPD). ßAR agonists induce the production of cAMP by adenylyl cyclases (ACs), activate PKA and cause bronchodilation. Several other G-protein coupled receptors (GPCR) expressed in human airway smooth muscle cells transduce extracellular signals through cAMP but these receptors elicit different cellular responses. Some G-protein coupled receptors couple to distinct adenylyl cyclases isoforms with different localization, partly explaining this compartmentation, but little is known about the downstream networks that result. We used quantitative phosphoproteomics to define the downstream signaling networks emanating from cAMP produced by two adenylyl cyclases isoforms with contrasting localization in uman airway smooth muscle. After a short stimulus of adenylyl cyclases activity using forskolin, phosphopeptides were analyzed by LC-MS/MS and differences between cells overexpressing AC2 (localized in non-raft membranes) or AC6 (localized in lipid raft membranes) were compared to control human airway smooth muscle. The degree of AC2 and AC6 overexpression was titrated to generate roughly equal forskolin-stimulated cAMP production. 14 Differentially phosphorylated proteins (DPPs) resulted from AC2 activity and 34 differentially phosphorylated proteins resulted from AC6 activity. Analysis of these hits with the STRING protein interaction tool showed that AC2 signaling is more associated with modifications in RNA/DNA binding proteins and microtubule/spindle body proteins while AC6 signaling is associated with proteins regulating autophagy, calcium-calmodulin (Ca2+/CaM) signaling, Rho GTPases and cytoskeletal regulation. One protein, OFD1, was regulated in opposite directions, with serine 899 phosphorylation increased in the AC6 condition 1.5-fold but decreased to 0.46-fold by AC2. In conclusion, quantitative phosphoproteomics is a powerful tool for deciphering the complex signaling networks resulting from discreet signaling events that occur in cAMP compartments. Our data show key differences in the cAMP pools generated from AC2 and AC6 activity and imply that distinct cellular responses are regulated by these two compartments.
Collapse
Affiliation(s)
- Isabella Cattani-Cavalieri
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, United States
| | - Yue Li
- Department of Pharmacy Practice, Chapman University School of Pharmacy, Irvine, CA, United States
| | - Jordyn Margolis
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, United States
| | - Amy Bogard
- AB Research LLC, Cincinnati, OH, United States
| | - Moom R. Roosan
- Department of Pharmacy Practice, Chapman University School of Pharmacy, Irvine, CA, United States
| | - Rennolds S. Ostrom
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, United States
| |
Collapse
|
5
|
Leavesley SJ, Johnson S, Paudel SS, Knighten J, Tambe DT, Francis M, Gong N, Taylor MS, Rich TC. Combined hyperspectral imaging, monolayer stress microscopy, and S8 image analysis approaches for simultaneously interrogating cellular signals and biomechanics. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2023; 12383:123830D. [PMID: 37051186 PMCID: PMC10084657 DOI: 10.1117/12.2650653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
Second messenger signals, e.g., Ca2+ and cyclic nucleotides, orchestrate a wide range of cellular events. The methods by which second messenger signals determine specific physiological responses are complex. Recent studies point to the importance of temporal and spatial encoding in determining signal specificity. Studies also indicate the importance of mechanical stimuli, substrate stiffness, and mechanical responses - the "mechanosome" - in regulating physiology. Hence, approaches that probe both chemical and mechanical signals are needed. Here, we report preliminary efforts to combine hyperspectral imaging for second messenger signal measurements, monolayer stress microscopy for mechanical force measurements, and S8 analysis software for quantifying localized signals - specifically, Ca2+ dynamics and mechanical forces in human airway smooth muscle cells (HASMCs). HASMCs were prepared as confluent monolayers on 11 kPa gels with embedded fluorescent microparticles that serve as fiducial markers as well as smaller microparticles to measure deformation (strain). Imaging was performed using a custom excitation-scanning hyperspectral microscope. Hyperspectral images were unmixed to identify signals from cellular fluorescent labels (e.g., CAL 590-AM) and fluorescent microparticles. Images were analyzed to quantify localized force dynamics through monolayer stress microscopy. S8 software was used to identify, track, and quantify spatially-localized Ca2+ activity. Results indicate that localized and transient cellular signals and forces can be quantified and mapped within cell populations. Importantly, these results establish a method for simultaneous interrogation of cellular signals and mechanical forces that may play synergistic roles in regulating downstream cellular physiology in confluent monolayers. This work was supported by NIH P01HL066299, R01HL137030, R01HL058506, and NSF MRI1725937. Drs. Leavesley and Rich disclose financial interest in a university start-up company, SpectraCyte LLC, to commercialize spectral imaging technologies.
Collapse
Affiliation(s)
- Silas J Leavesley
- Department of Chemical and Biomolecular Engineering
- Department of Pharmacology
- Center for Lung Biology
| | | | - Sunita S Paudel
- Center for Lung Biology
- Department of Physiology and Cell Biology
| | | | - Dhananjay T Tambe
- Department of Pharmacology
- Center for Lung Biology
- William B. Burnsed Jr. Department of Mechanical, Aerospace, and Biomedical Engineering
| | - Michael Francis
- Center for Lung Biology
- Department of Physiology and Cell Biology
| | - Na Gong
- Department of Electrical and Computer Engineering, University of South Alabama, Mobile, AL, USA 36688
| | - Mark S Taylor
- Center for Lung Biology
- Department of Physiology and Cell Biology
| | | |
Collapse
|
6
|
Bhadra A, Scruggs AK, Leavesley SJ, Annamdevula N, George AH, Britain AL, Francis CM, Knighten JM, Rich TC, Bauer NN. Extracellular vesicle-induced cyclic AMP signaling. Cell Signal 2022; 95:110348. [PMID: 35504529 PMCID: PMC10676271 DOI: 10.1016/j.cellsig.2022.110348] [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: 03/07/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 11/25/2022]
Abstract
Second messenger signaling is required for cellular processes. We previously reported that extracellular vesicles (EVs) from stimulated cultured endothelial cells contain the biochemical second messenger, cAMP. In the current study, we sought to determine whether cAMP-enriched EVs induce second messenger signaling pathways in naïve recipient cells. Our results indicate that cAMP-enriched EVs increase cAMP content sufficient to stimulate PKA activity. The implications of our work are that EVs represent a novel intercellular mechanism for second messenger, specifically cAMP, signaling.
Collapse
Affiliation(s)
- Aritra Bhadra
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL, United States of America; Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - April K Scruggs
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL, United States of America; Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Silas J Leavesley
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL, United States of America; Department of Chemical and Biomolecular Engineering, College of Engineering, University of South Alabama, Mobile, AL, United States of America; Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Naga Annamdevula
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL, United States of America; Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - April H George
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Andrea L Britain
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL, United States of America; Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Christopher M Francis
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL, United States of America; Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Jennifer M Knighten
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL, United States of America; Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Thomas C Rich
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL, United States of America; Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL, United States of America
| | - Natalie N Bauer
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL, United States of America; Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL, United States of America.
| |
Collapse
|
7
|
Ostrom KF, LaVigne JE, Brust TF, Seifert R, Dessauer CW, Watts VJ, Ostrom RS. Physiological roles of mammalian transmembrane adenylyl cyclase isoforms. Physiol Rev 2022; 102:815-857. [PMID: 34698552 PMCID: PMC8759965 DOI: 10.1152/physrev.00013.2021] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/20/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
Adenylyl cyclases (ACs) catalyze the conversion of ATP to the ubiquitous second messenger cAMP. Mammals possess nine isoforms of transmembrane ACs, dubbed AC1-9, that serve as major effector enzymes of G protein-coupled receptors (GPCRs). The transmembrane ACs display varying expression patterns across tissues, giving the potential for them to have a wide array of physiological roles. Cells express multiple AC isoforms, implying that ACs have redundant functions. Furthermore, all transmembrane ACs are activated by Gαs, so it was long assumed that all ACs are activated by Gαs-coupled GPCRs. AC isoforms partition to different microdomains of the plasma membrane and form prearranged signaling complexes with specific GPCRs that contribute to cAMP signaling compartments. This compartmentation allows for a diversity of cellular and physiological responses by enabling unique signaling events to be triggered by different pools of cAMP. Isoform-specific pharmacological activators or inhibitors are lacking for most ACs, making knockdown and overexpression the primary tools for examining the physiological roles of a given isoform. Much progress has been made in understanding the physiological effects mediated through individual transmembrane ACs. GPCR-AC-cAMP signaling pathways play significant roles in regulating functions of every cell and tissue, so understanding each AC isoform's role holds potential for uncovering new approaches for treating a vast array of pathophysiological conditions.
Collapse
Affiliation(s)
| | - Justin E LaVigne
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
| | - Tarsis F Brust
- Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, West Palm Beach, Florida
| | - Roland Seifert
- Institute of Pharmacology, Hannover Medical School, Hannover, Germany
| | - Carmen W Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas
| | - Val J Watts
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
- Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana
| | - Rennolds S Ostrom
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California
| |
Collapse
|
8
|
Anton SE, Kayser C, Maiellaro I, Nemec K, Möller J, Koschinski A, Zaccolo M, Annibale P, Falcke M, Lohse MJ, Bock A. Receptor-associated independent cAMP nanodomains mediate spatiotemporal specificity of GPCR signaling. Cell 2022; 185:1130-1142.e11. [PMID: 35294858 DOI: 10.1016/j.cell.2022.02.011] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 12/20/2021] [Accepted: 02/09/2022] [Indexed: 01/05/2023]
Abstract
G protein-coupled receptors (GPCRs) relay extracellular stimuli into specific cellular functions. Cells express many different GPCRs, but all these GPCRs signal to only a few second messengers such as cAMP. It is largely unknown how cells distinguish between signals triggered by different GPCRs to orchestrate their complex functions. Here, we demonstrate that individual GPCRs signal via receptor-associated independent cAMP nanodomains (RAINs) that constitute self-sufficient, independent cell signaling units. Low concentrations of glucagon-like peptide 1 (GLP-1) and isoproterenol exclusively generate highly localized cAMP pools around GLP-1- and β2-adrenergic receptors, respectively, which are protected from cAMP originating from other receptors and cell compartments. Mapping local cAMP concentrations with engineered GPCR nanorulers reveals gradients over only tens of nanometers that define the size of individual RAINs. The coexistence of many such RAINs allows a single cell to operate thousands of independent cellular signals simultaneously, rather than function as a simple "on/off" switch.
Collapse
Affiliation(s)
- Selma E Anton
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Strasse 10, 13125 Berlin, Germany; Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, 97078 Würzburg, Germany
| | - Charlotte Kayser
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | - Isabella Maiellaro
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, 97078 Würzburg, Germany; School of Life Sciences, Department of Neuroscience, University of Nottingham, Nottingham NG7 2UH, UK
| | - Katarina Nemec
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Strasse 10, 13125 Berlin, Germany; Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, 97078 Würzburg, Germany
| | - Jan Möller
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Strasse 10, 13125 Berlin, Germany; Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, 97078 Würzburg, Germany
| | - Andreas Koschinski
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | - Paolo Annibale
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Strasse 10, 13125 Berlin, Germany; Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, 97078 Würzburg, Germany; School of Physics and Astronomy, University of St Andrews, North Haugh, St. Andrews KY16 9SS, UK
| | - Martin Falcke
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Strasse 10, 13125 Berlin, Germany; Department of Physics, Humboldt University, Newtonstr. 15, 12489 Berlin, Germany
| | - Martin J Lohse
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Strasse 10, 13125 Berlin, Germany; Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, 97078 Würzburg, Germany; Institute for Chemistry and Biochemistry, Free University Berlin, Takustr. 3, 14195 Berlin, Germany; ISAR Bioscience Institute, Semmelweisstrasse 5, 82152 Planegg, Munich, Germany.
| | - Andreas Bock
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Strasse 10, 13125 Berlin, Germany; Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, 97078 Würzburg, Germany; Rudolf Boehm Institute of Pharmacology and Toxicology, Medical Faculty, University of Leipzig, Härtelstr. 16-18, 04107 Leipzig, Germany.
| |
Collapse
|
9
|
Rich TC, Xin W, Leavesley SJ, Francis CM, Taylor M. Ion Channel-Based Reporters for cAMP Detection. Methods Mol Biol 2022; 2483:265-279. [PMID: 35286682 DOI: 10.1007/978-1-0716-2245-2_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In the last 20 years tremendous progress has been made in the development of single cell cAMP sensors. Sensors are based upon cAMP binding proteins that have been modified to transduce cAMP concentrations into electrical or fluorescent readouts that can be readily detected using patch clamp amplifiers, photomultiplier tubes, or cameras. Here, we describe two complementary approaches for the detection and measurement of cAMP signals near the plasma membrane of cells using cyclic nucleotide (CNG) channel-based probes. These probes take advantage of the ability of CNG channels to transduce small changes in cAMP concentration into ionic flux through channel pores that can be readily detected by measuring Ca2+ and/or Mn2+ influx or by measuring ionic currents.
Collapse
Affiliation(s)
- Thomas C Rich
- Department of Pharmacology, University of South Alabama, Mobile, AL, USA.
- Center for Lung Biology, University of South Alabama, Mobile, AL, USA.
| | - Wenkuan Xin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Silas J Leavesley
- Department of Pharmacology, University of South Alabama, Mobile, AL, USA
- Center for Lung Biology, University of South Alabama, Mobile, AL, USA
- Department of Chemical and Biomolecular Engineering, University of South Alabama, Mobile, AL, USA
| | - C Michael Francis
- Center for Lung Biology, University of South Alabama, Mobile, AL, USA
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL, USA
| | - Mark Taylor
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL, USA
| |
Collapse
|
10
|
Agarwal SR, Sherpa RT, Moshal KS, Harvey RD. Compartmentalized cAMP signaling in cardiac ventricular myocytes. Cell Signal 2022; 89:110172. [PMID: 34687901 PMCID: PMC8602782 DOI: 10.1016/j.cellsig.2021.110172] [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] [Received: 08/31/2021] [Revised: 10/15/2021] [Accepted: 10/17/2021] [Indexed: 01/03/2023]
Abstract
Activation of different receptors that act by generating the common second messenger cyclic adenosine monophosphate (cAMP) can elicit distinct functional responses in cardiac myocytes. Selectively sequestering cAMP activity to discrete intracellular microdomains is considered essential for generating receptor-specific responses. The processes that control this aspect of compartmentalized cAMP signaling, however, are not completely clear. Over the years, technological innovations have provided critical breakthroughs in advancing our understanding of the mechanisms underlying cAMP compartmentation. Some of the factors identified include localized production of cAMP by differential distribution of receptors, localized breakdown of this second messenger by targeted distribution of phosphodiesterase enzymes, and limited diffusion of cAMP by protein kinase A (PKA)-dependent buffering or physically restricted barriers. The aim of this review is to provide a discussion of our current knowledge and highlight some of the gaps that still exist in the field of cAMP compartmentation in cardiac myocytes.
Collapse
|
11
|
Warren R, Rich T, Leavesley S, Phan AV. A three-dimensional finite element model of cAMP signals. FORCES IN MECHANICS 2021; 4. [PMID: 35072121 PMCID: PMC8773462 DOI: 10.1016/j.finmec.2021.100041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- R. Warren
- Department of Mechanical, Aerospace and Biomedical Engineering, University of South Alabama, Mobile, AL 36688, USA
| | - T.C. Rich
- Center for Lung Biology & Department of Pharmacology, University of South Alabama, Mobile, AL 36688, USA
| | - S.J. Leavesley
- Center for Lung Biology & Department of Pharmacology, University of South Alabama, Mobile, AL 36688, USA
- Department of Chemical and Biomolecular Engineering, University of South Alabama, Mobile, AL 36688, USA
| | - A.-V. Phan
- Department of Mechanical, Aerospace and Biomedical Engineering, University of South Alabama, Mobile, AL 36688, USA
- Corresponding author. (A.-V. Phan)
| |
Collapse
|
12
|
Wright PT, Gorelik J, Harding SE. Electrophysiological Remodeling: Cardiac T-Tubules and ß-Adrenoceptors. Cells 2021; 10:cells10092456. [PMID: 34572106 PMCID: PMC8468945 DOI: 10.3390/cells10092456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 01/09/2023] Open
Abstract
Beta-adrenoceptors (βAR) are often viewed as archetypal G-protein coupled receptors. Over the past fifteen years, investigations in cardiovascular biology have provided remarkable insights into this receptor family. These studies have shifted pharmacological dogma, from one which centralized the receptor to a new focus on structural micro-domains such as caveolae and t-tubules. Important studies have examined, separately, the structural compartmentation of ion channels and βAR. Despite links being assumed, relatively few studies have specifically examined the direct link between structural remodeling and electrical remodeling with a focus on βAR. In this review, we will examine the nature of receptor and ion channel dysfunction on a substrate of cardiomyocyte microdomain remodeling, as well as the likely ramifications for cardiac electrophysiology. We will then discuss the advances in methodologies in this area with a specific focus on super-resolution microscopy, fluorescent imaging, and new approaches involving microdomain specific, polymer-based agonists. The advent of powerful computational modelling approaches has allowed the science to shift from purely empirical work, and may allow future investigations based on prediction. Issues such as the cross-reactivity of receptors and cellular heterogeneity will also be discussed. Finally, we will speculate as to the potential developments within this field over the next ten years.
Collapse
Affiliation(s)
- Peter T. Wright
- School of Life & Health Sciences, University of Roehampton, Holybourne Avenue, London SW15 4JD, UK;
- Cardiac Section, National Heart and Lung Institute (NHLI), Faculty of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK;
| | - Julia Gorelik
- Cardiac Section, National Heart and Lung Institute (NHLI), Faculty of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK;
| | - Sian E. Harding
- Cardiac Section, National Heart and Lung Institute (NHLI), Faculty of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK;
- Correspondence:
| |
Collapse
|
13
|
Harvey RD, Clancy CE. Mechanisms of cAMP compartmentation in cardiac myocytes: experimental and computational approaches to understanding. J Physiol 2021; 599:4527-4544. [PMID: 34510451 DOI: 10.1113/jp280801] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/07/2021] [Indexed: 01/04/2023] Open
Abstract
The small diffusible second messenger 3',5'-cyclic adenosine monophosphate (cAMP) is found in virtually every cell in our bodies, where it mediates responses to a variety of different G protein coupled receptors (GPCRs). In the heart, cAMP plays a critical role in regulating many different aspects of cardiac myocyte function, including gene transcription, cell metabolism, and excitation-contraction coupling. Yet, not all GPCRs that stimulate cAMP production elicit the same responses. Subcellular compartmentation of cAMP is essential to explain how different receptors can utilize the same diffusible second messenger to elicit unique functional responses. However, the mechanisms contributing to this behaviour and its significance in producing physiological and pathological responses are incompletely understood. Mathematical modelling has played an essential role in gaining insight into these questions. This review discusses what we currently know about cAMP compartmentation in cardiac myocytes and questions that are yet to be answered.
Collapse
Affiliation(s)
- Robert D Harvey
- Department of Pharmacology, University of Nevada, Reno, NV, 89557, USA
| | - Colleen E Clancy
- Department of Physiology and Membrane Biology, University of California-Davis, Davis, CA, 95616, USA
| |
Collapse
|
14
|
Zhang X, Pizzoni A, Hong K, Naim N, Qi C, Korkhov V, Altschuler DL. CAP1 binds and activates adenylyl cyclase in mammalian cells. Proc Natl Acad Sci U S A 2021; 118:e2024576118. [PMID: 34099549 PMCID: PMC8214675 DOI: 10.1073/pnas.2024576118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
CAP1 (Cyclase-Associated Protein 1) is highly conserved in evolution. Originally identified in yeast as a bifunctional protein involved in Ras-adenylyl cyclase and F-actin dynamics regulation, the adenylyl cyclase component seems to be lost in mammalian cells. Prompted by our recent identification of the Ras-like small GTPase Rap1 as a GTP-independent but geranylgeranyl-specific partner for CAP1, we hypothesized that CAP1-Rap1, similar to CAP-Ras-cyclase in yeast, might play a critical role in cAMP dynamics in mammalian cells. In this study, we report that CAP1 binds and activates mammalian adenylyl cyclase in vitro, modulates cAMP in live cells in a Rap1-dependent manner, and affects cAMP-dependent proliferation. Utilizing deletion and mutagenesis approaches, we mapped the interaction of CAP1-cyclase with CAP's N-terminal domain involving critical leucine residues in the conserved RLE motifs and adenylyl cyclase's conserved catalytic loops (e.g., C1a and/or C2a). When combined with a FRET-based cAMP sensor, CAP1 overexpression-knockdown strategies, and the use of constitutively active and negative regulators of Rap1, our studies highlight a critical role for CAP1-Rap1 in adenylyl cyclase regulation in live cells. Similarly, we show that CAP1 modulation significantly affected cAMP-mediated proliferation in an RLE motif-dependent manner. The combined study indicates that CAP1-cyclase-Rap1 represents a regulatory unit in cAMP dynamics and biology. Since Rap1 is an established downstream effector of cAMP, we advance the hypothesis that CAP1-cyclase-Rap1 represents a positive feedback loop that might be involved in cAMP microdomain establishment and localized signaling.
Collapse
Affiliation(s)
- Xuefeng Zhang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Alejandro Pizzoni
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Kyoungja Hong
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Nyla Naim
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Chao Qi
- Institute of Molecular Biology and Biophysics, Paul Scherrer Institute, 5232 Villigen PSI, Switzerland
| | - Volodymyr Korkhov
- Institute of Molecular Biology and Biophysics, Paul Scherrer Institute, 5232 Villigen PSI, Switzerland
| | - Daniel L Altschuler
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261;
| |
Collapse
|
15
|
McCabe KJ, Rangamani P. Computational modeling approaches to cAMP/PKA signaling in cardiomyocytes. J Mol Cell Cardiol 2021; 154:32-40. [PMID: 33548239 DOI: 10.1016/j.yjmcc.2021.01.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/11/2021] [Accepted: 01/15/2021] [Indexed: 12/12/2022]
Abstract
The cAMP/PKA pathway is a fundamental regulator of excitation-contraction coupling in cardiomyocytes. Activation of cAMP has a variety of downstream effects on cardiac function including enhanced contraction, accelerated relaxation, adaptive stress response, mitochondrial regulation, and gene transcription. Experimental advances have shed light on the compartmentation of cAMP and PKA, which allow for control over the varied targets of these second messengers and is disrupted in heart failure conditions. Computational modeling is an important tool for understanding the spatial and temporal complexities of this system. In this review article, we outline the advances in computational modeling that have allowed for deeper understanding of cAMP/PKA dynamics in the cardiomyocyte in health and disease, and explore new modeling frameworks that may bring us closer to a more complete understanding of this system. We outline various compartmental and spatial signaling models that have been used to understand how β-adrenergic signaling pathways function in a variety of simulation conditions. We also discuss newer subcellular models of cardiovascular function that may be used as templates for the next phase of computational study of cAMP and PKA in the heart, and outline open challenges which are important to consider in future models.
Collapse
Affiliation(s)
- Kimberly J McCabe
- Simula Research Laboratory, Department of Computational Physiology, PO Box 134, 1325 Lysaker, Norway.
| | - Padmini Rangamani
- University of California San Diego, Department of Mechanical and Aerospace Engineering, 9500 Gilman Drive MC 0411, La Jolla, CA 92093, United States of America
| |
Collapse
|
16
|
Abstract
The field of cAMP signaling is witnessing exciting developments with the recognition that cAMP is compartmentalized and that spatial regulation of cAMP is critical for faithful signal coding. This realization has changed our understanding of cAMP signaling from a model in which cAMP connects a receptor at the plasma membrane to an intracellular effector in a linear pathway to a model in which cAMP signals propagate within a complex network of alternative branches and the specific functional outcome strictly depends on local regulation of cAMP levels and on selective activation of a limited number of branches within the network. In this review, we cover some of the early studies and summarize more recent evidence supporting the model of compartmentalized cAMP signaling, and we discuss how this knowledge is starting to provide original mechanistic insight into cell physiology and a novel framework for the identification of disease mechanisms that potentially opens new avenues for therapeutic interventions. SIGNIFICANCE STATEMENT: cAMP mediates the intracellular response to multiple hormones and neurotransmitters. Signal fidelity and accurate coordination of a plethora of different cellular functions is achieved via organization of multiprotein signalosomes and cAMP compartmentalization in subcellular nanodomains. Defining the organization and regulation of subcellular cAMP nanocompartments is necessary if we want to understand the complex functional ramifications of pharmacological treatments that target G protein-coupled receptors and for generating a blueprint that can be used to develop precision medicine interventions.
Collapse
Affiliation(s)
- Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Anna Zerio
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Miguel J Lobo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
17
|
Thomas M, Hughes TE. Optically activated, customizable, excitable cells. PLoS One 2020; 15:e0229051. [PMID: 33378334 PMCID: PMC7773186 DOI: 10.1371/journal.pone.0229051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 12/03/2020] [Indexed: 11/29/2022] Open
Abstract
Genetically encoded fluorescent biosensors are powerful tools for studying complex signaling in the nervous system, and now both Ca2+ and voltage sensors are available to study the signaling behavior of entire neural circuits. There is a pressing need for improved sensors, but improving them is challenging because testing them involves a low throughput, labor-intensive processes. Our goal was to create synthetic, excitable cells that can be activated with brief pulses of blue light and serve as a medium throughput platform for screening the next generation of sensors. In this live cell system, blue light activates an adenylyl cyclase enzyme (bPAC) that increases intracellular cAMP (Stierl M et al. 2011). In turn, the cAMP opens a cAMP-gated ion channel. This produces slow, whole-cell Ca2+ transients and voltage changes. To increase the speed of these transients, we add the inwardly rectifying potassium channel Kir2.1, the bacterial voltage-gated sodium channel NAVROSD, and Connexin-43. The result is a highly reproducible, medium-throughput, live cell system that can be used to screen voltage and Ca2+ sensors.
Collapse
Affiliation(s)
- Merrilee Thomas
- Department of Neuroscience and Cell Biology, Montana State University, Bozeman, Montana, United States of America
| | - Thomas E. Hughes
- Department of Neuroscience and Cell Biology, Montana State University, Bozeman, Montana, United States of America
- Montana Molecular, Bozeman, Montana, United States of America
| |
Collapse
|
18
|
Zhang JZ, Lu TW, Stolerman LM, Tenner B, Yang JR, Zhang JF, Falcke M, Rangamani P, Taylor SS, Mehta S, Zhang J. Phase Separation of a PKA Regulatory Subunit Controls cAMP Compartmentation and Oncogenic Signaling. Cell 2020; 182:1531-1544.e15. [PMID: 32846158 PMCID: PMC7502557 DOI: 10.1016/j.cell.2020.07.043] [Citation(s) in RCA: 203] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 03/30/2020] [Accepted: 07/30/2020] [Indexed: 12/25/2022]
Abstract
The fidelity of intracellular signaling hinges on the organization of dynamic activity architectures. Spatial compartmentation was first proposed over 30 years ago to explain how diverse G protein-coupled receptors achieve specificity despite converging on a ubiquitous messenger, cyclic adenosine monophosphate (cAMP). However, the mechanisms responsible for spatially constraining this diffusible messenger remain elusive. Here, we reveal that the type I regulatory subunit of cAMP-dependent protein kinase (PKA), RIα, undergoes liquid-liquid phase separation (LLPS) as a function of cAMP signaling to form biomolecular condensates enriched in cAMP and PKA activity, critical for effective cAMP compartmentation. We further show that a PKA fusion oncoprotein associated with an atypical liver cancer potently blocks RIα LLPS and induces aberrant cAMP signaling. Loss of RIα LLPS in normal cells increases cell proliferation and induces cell transformation. Our work reveals LLPS as a principal organizer of signaling compartments and highlights the pathological consequences of dysregulating this activity architecture.
Collapse
Affiliation(s)
- Jason Z Zhang
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Tsan-Wen Lu
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Lucas M Stolerman
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Brian Tenner
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jessica R Yang
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jin-Fan Zhang
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Martin Falcke
- Mathematical Cell Physiology, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany; Department of Physics, Humboldt University, 12489 Berlin, Germany
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Susan S Taylor
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA; Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sohum Mehta
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jin Zhang
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
19
|
Bang J, Zippin JH. Cyclic adenosine monophosphate (cAMP) signaling in melanocyte pigmentation and melanomagenesis. Pigment Cell Melanoma Res 2020; 34:28-43. [PMID: 32777162 DOI: 10.1111/pcmr.12920] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/24/2020] [Accepted: 08/07/2020] [Indexed: 12/12/2022]
Abstract
The second messenger cyclic adenosine monophosphate (cAMP) regulates numerous functions in both benign melanocytes and melanoma cells. cAMP is generated from two distinct sources, transmembrane and soluble adenylyl cyclases (tmAC and sAC, respectively), and is degraded by a family of proteins called phosphodiesterases (PDEs). cAMP signaling can be regulated in many different ways and can lead to varied effects in melanocytes. It was recently revealed that distinct cAMP signaling pathways regulate pigmentation by either altering pigment gene expression or the pH of melanosomes. In the context of melanoma, many studies report seemingly contradictory roles for cAMP in tumorigenesis. For example, cAMP signaling has been implicated in both cancer promotion and suppression, as well as both therapy resistance and sensitization. This conundrum in the field may be explained by the fact that cAMP signals in discrete microdomains and each microdomain can mediate differential cellular functions. Here, we review the role of cAMP signaling microdomains in benign melanocyte biology, focusing on pigmentation, and in melanomagenesis.
Collapse
Affiliation(s)
- Jakyung Bang
- Department of Dermatology, Joan and Sanford I. Weill Medical College of Cornell University, New York, NY, USA
| | - Jonathan H Zippin
- Department of Dermatology, Joan and Sanford I. Weill Medical College of Cornell University, New York, NY, USA
| |
Collapse
|
20
|
Bock A, Annibale P, Konrad C, Hannawacker A, Anton SE, Maiellaro I, Zabel U, Sivaramakrishnan S, Falcke M, Lohse MJ. Optical Mapping of cAMP Signaling at the Nanometer Scale. Cell 2020; 182:1519-1530.e17. [PMID: 32846156 DOI: 10.1016/j.cell.2020.07.035] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 03/30/2020] [Accepted: 07/23/2020] [Indexed: 10/23/2022]
Abstract
Cells relay a plethora of extracellular signals to specific cellular responses by using only a few second messengers, such as cAMP. To explain signaling specificity, cAMP-degrading phosphodiesterases (PDEs) have been suggested to confine cAMP to distinct cellular compartments. However, measured rates of fast cAMP diffusion and slow PDE activity render cAMP compartmentalization essentially impossible. Using fluorescence spectroscopy, we show that, contrary to earlier data, cAMP at physiological concentrations is predominantly bound to cAMP binding sites and, thus, immobile. Binding and unbinding results in largely reduced cAMP dynamics, which we term "buffered diffusion." With a large fraction of cAMP being buffered, PDEs can create nanometer-size domains of low cAMP concentrations. Using FRET-cAMP nanorulers, we directly map cAMP gradients at the nanoscale around PDE molecules and the areas of resulting downstream activation of cAMP-dependent protein kinase (PKA). Our study reveals that spatiotemporal cAMP signaling is under precise control of nanometer-size domains shaped by PDEs that gate activation of downstream effectors.
Collapse
Affiliation(s)
- Andreas Bock
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Str. 10, 13125 Berlin, Germany; Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, 97078 Würzburg, Germany.
| | - Paolo Annibale
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Str. 10, 13125 Berlin, Germany; Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, 97078 Würzburg, Germany
| | - Charlotte Konrad
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Str. 10, 13125 Berlin, Germany; Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, 97078 Würzburg, Germany
| | - Annette Hannawacker
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, 97078 Würzburg, Germany
| | - Selma E Anton
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Str. 10, 13125 Berlin, Germany; Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, 97078 Würzburg, Germany
| | - Isabella Maiellaro
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, 97078 Würzburg, Germany
| | - Ulrike Zabel
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, 97078 Würzburg, Germany
| | - Sivaraj Sivaramakrishnan
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Martin Falcke
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Str. 10, 13125 Berlin, Germany; Department of Physics, Humboldt University, Newtonstr. 15, 12489 Berlin, Germany
| | - Martin J Lohse
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Str. 10, 13125 Berlin, Germany; Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, 97078 Würzburg, Germany; Institute for Chemistry and Biochemistry, Free University, Takustr. 3, 14195 Berlin, Germany; ISAR Bioscience Institute, 82152 Munich/Planegg, Germany.
| |
Collapse
|
21
|
Wang X, Cai B, Yang X, Sonubi OO, Zheng Z, Ramakrishnan R, Shi H, Valenti L, Pajvani UB, Sandhu J, Infante RE, Radhakrishnan A, Covey DF, Guan KL, Buck J, Levin LR, Tontonoz P, Schwabe RF, Tabas I. Cholesterol Stabilizes TAZ in Hepatocytes to Promote Experimental Non-alcoholic Steatohepatitis. Cell Metab 2020; 31:969-986.e7. [PMID: 32259482 PMCID: PMC7313619 DOI: 10.1016/j.cmet.2020.03.010] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 01/04/2020] [Accepted: 03/11/2020] [Indexed: 12/15/2022]
Abstract
Incomplete understanding of how hepatosteatosis transitions to fibrotic non-alcoholic steatohepatitis (NASH) has limited therapeutic options. Two molecules that are elevated in hepatocytes in human NASH liver are cholesterol, whose mechanistic link to NASH remains incompletely understood, and TAZ, a transcriptional regulator that promotes fibrosis but whose mechanism of increase in NASH is unknown. We now show that increased hepatocyte cholesterol upregulates TAZ and promotes fibrotic NASH. ASTER-B/C-mediated internalization of plasma membrane cholesterol activates soluble adenylyl cyclase (sAC; ADCY10), triggering a calcium-RhoA-mediated pathway that suppresses β-TrCP/proteasome-mediated TAZ degradation. In mice fed with a cholesterol-rich NASH-inducing diet, hepatocyte-specific silencing of ASTER-B/C, sAC, or RhoA decreased TAZ and ameliorated fibrotic NASH. The cholesterol-TAZ pathway is present in primary human hepatocytes, and associations among liver cholesterol, TAZ, and RhoA in human NASH liver are consistent with the pathway. Thus, hepatocyte cholesterol contributes to fibrotic NASH by increasing TAZ, suggesting new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA.
| | - Bishuang Cai
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Xiaoming Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, PRC
| | - Oluwatoni O Sonubi
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Ze Zheng
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Rajasekhar Ramakrishnan
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Hongxue Shi
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milano 20122, Italy; Translational Medicine - Transfusion Medicine and Hematology, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Utpal B Pajvani
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jaspreet Sandhu
- Department of Pathology and Laboratory Medicine, Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90272, USA
| | - Rodney E Infante
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Arun Radhakrishnan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Douglas F Covey
- Department of Developmental Biology and Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jochen Buck
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Lonny R Levin
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90272, USA
| | - Robert F Schwabe
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Institute of Human Nutrition, Columbia University, New York, NY 10032, USA
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
22
|
Beute J, Ganesh K, Nastiti H, Hoogenboom R, Bos V, Folkerts J, Schreurs MWJ, Hockman S, Hendriks RW, KleinJan A. PDE3 Inhibition Reduces Epithelial Mast Cell Numbers in Allergic Airway Inflammation and Attenuates Degranulation of Basophils and Mast Cells. Front Pharmacol 2020; 11:470. [PMID: 32425769 PMCID: PMC7206980 DOI: 10.3389/fphar.2020.00470] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 03/25/2020] [Indexed: 11/13/2022] Open
Abstract
Epithelial mast cells are generally present in the airways of patients with allergic asthma that are inadequately controlled. Airway mast cells (MCs) are critically involved in allergic airway inflammation and contribute directly to the main symptoms of allergic patients. Phosphodiesterase 3 (PDE3) tailors signaling of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP), which are critical intracellular second messenger molecules in various signaling pathways. This paper investigates the pathophysiological role and disease-modifying effects of PDE3 in mouse bone marrow-derived MCs (bmMCs), human LAD2- and HMC1 mast cell lines, human blood basophils, and peripheral blood-derived primary human MCs (HuMCs). In a chronic house dust mite (HDM)-driven allergic airway inflammation mouse model, we observed that PDE3 deficiency or PDE3 inhibition (PDE3i) therapy reduced the numbers of epithelial MCs, when compared to control mice. Mouse bone marrow-derived MCs (bmMCs) and the human HMC1 and LAD2 cell lines predominantly expressed PDE3B and PDE4A. BmMCs from Pde3−/− mice showed reduced loss of the degranulation marker CD107b compared with wild-type BmMCs, when stimulated in an immunoglobulin E (IgE)-dependent manner. Following both IgE-mediated and substance P-mediated activation, PDE3i-pretreated basophils, LAD2 cells, and HuMCs, showed less degranulation than diluent controls, as measured by surface CD63 expression. MCs lacking PDE3 or treated with the PDE3i enoximone exhibited a lower calcium flux upon stimulation with ionomycine. In conclusion PDE3 plays a critical role in basophil and mast cell degranulation and therefore its inhibition may be a treatment option in allergic disease.
Collapse
Affiliation(s)
- Jan Beute
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Keerthana Ganesh
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Hedwika Nastiti
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Robin Hoogenboom
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Vivica Bos
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Jelle Folkerts
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| | | | - Steve Hockman
- Flow Cytometry Core of the National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, United States
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Alex KleinJan
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
23
|
Schmidt M, Cattani-Cavalieri I, Nuñez FJ, Ostrom RS. Phosphodiesterase isoforms and cAMP compartments in the development of new therapies for obstructive pulmonary diseases. Curr Opin Pharmacol 2020; 51:34-42. [PMID: 32622335 PMCID: PMC7529846 DOI: 10.1016/j.coph.2020.05.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/17/2020] [Accepted: 05/22/2020] [Indexed: 12/20/2022]
Abstract
The second messenger molecule 3'5'-cyclic adenosine monophosphate (cAMP) imparts several beneficial effects in lung diseases such as asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). While cAMP is bronchodilatory in asthma and COPD, it also displays anti-fibrotic properties that limit fibrosis. Phosphodiesterases (PDEs) metabolize cAMP and thus regulate cAMP signaling. While some existing therapies inhibit PDEs, there are only broad family specific inhibitors. The understanding of cAMP signaling compartments, some centered around lipid rafts/caveolae, has led to interest in defining how specific PDE isoforms maintain these signaling microdomains. The possible altered expression of PDEs, and thus abnormal cAMP signaling, in obstructive lung diseases has been poorly explored. We propose that inhibition of specific PDE isoforms can improve therapy of obstructive lung diseases by amplifying specific cAMP signals in discreet microdomains.
Collapse
Affiliation(s)
- Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, GRIAC, Groningen, The Netherlands
| | - Isabella Cattani-Cavalieri
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, GRIAC, Groningen, The Netherlands; Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Francisco J Nuñez
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA
| | - Rennolds S Ostrom
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, USA.
| |
Collapse
|
24
|
Nanometric targeting of type 9 adenylyl cyclase in heart. Biochem Soc Trans 2020; 47:1749-1756. [PMID: 31769471 DOI: 10.1042/bst20190227] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 12/15/2022]
Abstract
Adenylyl cyclases (ACs) convert ATP into the classical second messenger cyclic adenosine monophosphate (cAMP). Cardiac ACs, specifically AC5, AC6, and AC9, regulate cAMP signaling controlling functional outcomes such as heart rate, contractility and relaxation, gene regulation, stress responses, and glucose and lipid metabolism. With so many distinct functional outcomes for a single second messenger, the cell creates local domains of cAMP signaling to correctly relay signals. Targeting of ACs to A-kinase anchoring proteins (AKAPs) not only localizes ACs, but also places them within signaling nanodomains, where cAMP levels and effects can be highly regulated. Here we will discuss the recent work on the structure, regulation and physiological functions of AC9 in the heart, where it accounts for <3% of total AC activity. Despite the small contribution of AC9 to total cardiac cAMP production, AC9 binds and regulates local PKA phosphorylation of Yotiao-IKs and Hsp20, demonstrating a role for nanometric targeting of AC9.
Collapse
|
25
|
Deal J, Annamdevula N, Pleshinger DJ, Griswold JR, Odom A, Tayara A, Lall M, Browning C, Parker M, Rich TC, Leavesley SJ. Comparison of spectral FRET microscopy approaches for single-cell analysis. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2020; 11243:112430Y. [PMID: 34035557 PMCID: PMC8142325 DOI: 10.1117/12.2546308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Förster resonance energy transfer (FRET) is a valuable tool for measuring molecular distances and the effects of biological processes such as cyclic nucleotide messenger signaling and protein localization. Most FRET techniques require two fluorescent proteins with overlapping excitation/emission spectral pairing to maximize detection sensitivity and FRET efficiency. FRET microscopy often utilizes differing peak intensities of the selected fluorophores measured through different optical filter sets to estimate the FRET index or efficiency. Microscopy platforms used to make these measurements include wide-field, laser scanning confocal, and fluorescence lifetime imaging. Each platform has associated advantages and disadvantages, such as speed, sensitivity, specificity, out-of-focus fluorescence, and Z-resolution. In this study, we report comparisons among multiple microscopy and spectral filtering platforms such as standard 2-filter FRET, emission-scanning hyperspectral imaging, and excitation-scanning hyperspectral imaging. Samples of human embryonic kidney (HEK293) cells were grown on laminin-coated 28 mm round gridded glass coverslips (10816, Ibidi, Fitchburg, Wisconsin) and transfected with adenovirus encoding a cAMP-sensing FRET probe composed of a FRET donor (Turquoise) and acceptor (Venus). Additionally, 3 FRET "controls" with fixed linker lengths between Turquoise and Venus proteins were used for inter-platform validation. Grid locations were logged, recorded with light micrographs, and used to ensure that whole-cell FRET was compared on a cell-by-cell basis among the different microscopy platforms. FRET efficiencies were also calculated and compared for each method. Preliminary results indicate that hyperspectral methods increase the signal-to-noise ratio compared to a standard 2-filter approach.
Collapse
Affiliation(s)
- Joshua Deal
- Department of Chemical & Biomolecular Engineering, University of South Alabama
- Center for Lung Biology, University of South Alabama
- Department of Pharmacology, University of South Alabama
| | - Naga Annamdevula
- Center for Lung Biology, University of South Alabama
- Department of Pharmacology, University of South Alabama
| | - Donald John Pleshinger
- Center for Lung Biology, University of South Alabama
- Department of Pharmacology, University of South Alabama
| | | | - Aliyah Odom
- Department of Chemical & Biomolecular Engineering, University of South Alabama
| | - Alia Tayara
- Department of Chemical & Biomolecular Engineering, University of South Alabama
| | - Malvika Lall
- College of Medicine, University of South Alabama
| | - Craig Browning
- Department of Chemical & Biomolecular Engineering, University of South Alabama
- Systems Engineering, University of South Alabama
| | - Marina Parker
- Department of Chemical & Biomolecular Engineering, University of South Alabama
- Systems Engineering, University of South Alabama
| | - Thomas C Rich
- Center for Lung Biology, University of South Alabama
- Department of Pharmacology, University of South Alabama
| | - Silas J Leavesley
- Department of Chemical & Biomolecular Engineering, University of South Alabama
- Center for Lung Biology, University of South Alabama
- Department of Pharmacology, University of South Alabama
| |
Collapse
|
26
|
Seth A, Finkbeiner M, Grischin J, Schultz JE. Gsα stimulation of mammalian adenylate cyclases regulated by their hexahelical membrane anchors. Cell Signal 2020; 68:109538. [PMID: 31931092 DOI: 10.1016/j.cellsig.2020.109538] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 01/09/2020] [Accepted: 01/09/2020] [Indexed: 12/14/2022]
Abstract
Mammalian adenylate cyclases (ACs) are pseudoheterodimers with dissimilar hexahelical membrane-anchors, isoform-specifically conserved for more than half a billion years. We exchanged both membrane anchors of the AC isoform 2 by the quorum-sensing receptor from Vibrio harveyi, CqsS, which has a ligand, Cholera-Autoinducer-1 (CAI-1). In the chimera, AC activity was stimulated by Gsα, CAI-1 had no effect. Surprisingly, CAI-1 inhibited Gsα stimulation. We report that Gsα stimulation of human AC isoforms 2, 3, 5, and 9 expressed in Sf9 cells is inhibited by serum as is AC activity in membranes isolated from rat brain cortex. AC2 activation by forskolin or forskolin/Gsα was similarly inhibited. Obviously, serum contains as yet unidentified factors affecting AC activity. The data establish a linkage in ACs, in which the membrane anchors, as receptors, transduce extracellular signals to the cytosolic catalytic dimer. A mechanistic three state model of AC regulation is presented compatible with all known regulatory inputs into mammalian ACs. The data allow designating the membrane anchors of mammalian ACs as orphan receptors, and establish a new level of AC regulation.
Collapse
Affiliation(s)
- Anubha Seth
- Pharmazeutisches Institut der Universität Tübingen, Tübingen, Germany; Max-Planck-Institut für Entwicklungsbiologie, Tübingen, Germany
| | | | - Julia Grischin
- Max-Planck-Institut für Entwicklungsbiologie, Tübingen, Germany
| | - Joachim E Schultz
- Pharmazeutisches Institut der Universität Tübingen, Tübingen, Germany.
| |
Collapse
|
27
|
Naim N, Reece JM, Zhang X, Altschuler DL. Dual Activation of cAMP Production Through Photostimulation or Chemical Stimulation. Methods Mol Biol 2020; 2173:201-216. [PMID: 32651920 PMCID: PMC7968876 DOI: 10.1007/978-1-0716-0755-8_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
cAMP is a crucial mediator of multiple cell signaling pathways. This cyclic nucleotide requires strict spatiotemporal control for effective function. Light-activated proteins have become a powerful tool to study signaling kinetics due to having quick on/off rates and minimal off-target effects. The photoactivated adenylyl cyclase from Beggiatoa (bPAC) produces cAMP rapidly upon stimulation with blue light. However, light delivery is not always feasible, especially in vivo. Hence, we created a luminescence-activated cyclase by fusing bPAC with nanoluciferase (nLuc) to allow chemical activation of cAMP activity. This dual-activated adenylyl cyclase can be stimulated using short bursts of light or long-term chemical activation with furimazine and other related luciferins. Together these can be used to mimic transient, chronic, and oscillating patterns of cAMP signaling. Moreover, when coupled to compartment-specific targeting domains, these reagents provide a new powerful tool for cAMP spatiotemporal dynamic studies. Here, we describe detailed methods for working with bPAC-nLuc in mammalian cells, stimulating cAMP production with light and luciferins, and measuring total cAMP accumulation.
Collapse
Affiliation(s)
- Nyla Naim
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Molecular Pharmacology Training Program, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology, Addgene, Watertown, MA, USA
| | - Jeff M Reece
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Advanced Light Microscopy & Image Analysis Core (ALMIAC), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Bethesda, MD, USA
| | - Xuefeng Zhang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel L Altschuler
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
28
|
Stone N, Shettlesworth S, Rich TC, Leavesley SJ, Phan AV. A two-dimensional finite element model of cyclic adenosine monophosphate (cAMP) intracellular signaling. SN APPLIED SCIENCES 2019; 1. [PMID: 33615142 DOI: 10.1007/s42452-019-1757-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
In this work, we present a two-dimensional finite element analysis (FEA) model that describes fundamental intracellular signals of cyclic adenosine monophosphate (cAMP) in a general fashion. The model was subsequently solved numerically and the results were displayed in forms of time-course plots of cAMP concentration at a cellular location or color-filled contour maps of cAMP signal distribution within the cell at specific time points. Basic intracellular cAMP signaling was described in this model so it can be numerically validated by verifying its numerical results against available analytical solutions and against results obtained from other numerical techniques reported in the literature. This is the first important step before the model can be expanded in future work. Model simulations demonstrate that under certain conditions, sustained cAMP concentrations can be formed within endothelial cells (ECs), similar to those observed in rat pulmonary microvascular ECs. Spatial and temporal cAMP dynamic simulations indicated that the proposed FEA model is an effective tool for the study of the kinetics and spatial spread of second messenger signaling and can be expanded to simulate second messenger signals in the pulmonary vasculature.
Collapse
Affiliation(s)
- N Stone
- William B. Burnsed, Jr. Department of Mechanical Engineering, University of South Alabama, Mobile, AL 36688, USA
| | - S Shettlesworth
- William B. Burnsed, Jr. Department of Mechanical Engineering, University of South Alabama, Mobile, AL 36688, USA
| | - T C Rich
- Center for Lung Biology & Department of Pharmacology, University of South Alabama, Mobile, AL 36688, USA
| | - S J Leavesley
- Department of Chemical and Biomolecular Engineering & Department of Pharmacology, University of South Alabama, Mobile, AL 36688, USA
| | - A-V Phan
- William B. Burnsed, Jr. Department of Mechanical Engineering, University of South Alabama, Mobile, AL 36688, USA
| |
Collapse
|
29
|
Ohadi D, Rangamani P. Geometric Control of Frequency Modulation of cAMP Oscillations due to Calcium in Dendritic Spines. Biophys J 2019; 117:1981-1994. [PMID: 31668747 PMCID: PMC7018999 DOI: 10.1016/j.bpj.2019.10.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 07/30/2019] [Accepted: 10/02/2019] [Indexed: 12/12/2022] Open
Abstract
The spatiotemporal regulation of cyclic adenosine monophosphate (cAMP) and its dynamic interactions with other second messengers such as calcium are critical features of signaling specificity required for neuronal development and connectivity. cAMP is known to contribute to long-term potentiation and memory formation by controlling the formation and regulation of dendritic spines. Despite the recent advances in biosensing techniques for monitoring spatiotemporal cAMP dynamics, the underlying molecular mechanisms that attribute to the subcellular modulation of cAMP remain unknown. In this work, we model the spatiotemporal dynamics of calcium-induced cAMP signaling pathway in dendritic spines. Using a three-dimensional reaction-diffusion model, we investigate the effect of different spatial characteristics of cAMP dynamics that may be responsible for subcellular regulation of cAMP concentrations. Our model predicts that the volume/surface ratio of the spine, regulated through the spine head size, spine neck size, and the presence of physical barriers (spine apparatus), is an important regulator of cAMP dynamics. Furthermore, localization of the enzymes responsible for the synthesis and degradation of cAMP in different compartments also modulates the oscillatory patterns of cAMP through exponential relationships. Our findings shed light on the significance of complex geometric and localization relationships for cAMP dynamics in dendritic spines.
Collapse
Affiliation(s)
- Donya Ohadi
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, California
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, California.
| |
Collapse
|
30
|
Rich TC, Griswold JR, Deal J, Annamdevula N, McAlister K, Mayes S, Browning C, Parker M, Leavelsey SJ. Hyperspectral imaging microscopy for measurement of localized second messenger signals in single cells. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2019; 10881:108811F. [PMID: 34045781 PMCID: PMC8151147 DOI: 10.1117/12.2508052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Ca2+ and cAMP are ubiquitous second messengers known to differentially regulate a variety of cellular functions over a wide range of timescales. Studies from a variety of groups support the hypothesis that these signals can be localized to discrete locations within cells, and that this subcellular localization is a critical component of signaling specificity. However, to date, it has been difficult to track second messenger signals at multiple locations within a single cell. This difficulty is largely due to the inability to measure multiplexed florescence signals in real time. To overcome this limitation, we have utilized both emission scan- and excitation scan-based hyperspectral imaging approaches to track second messenger signals as well as labeled cellular structures and/or proteins in the same cell. We have previously reported that hyperspectral imaging techniques improve the signal-to-noise ratios of both fluorescence and FRET measurements, and are thus well suited for the measurement of localized second messenger signals. Using these approaches, we have measured near plasma membrane and near nuclear membrane cAMP signals, as well as distributed signals within the cytosol, in several cell types including airway smooth muscle, pulmonary endothelial, and HEK-293 cells. We have also measured cAMP and Ca2+ signals near autofluorescent structures that appear to be golgi. Our data demonstrate that hyperspectral imaging approaches provide unique insight into the spatial and kinetic distributions of cAMP and Ca2+ signals in single cells.
Collapse
Affiliation(s)
- Thomas C Rich
- Pharmacology, University of South Alabama, AL 36688
- Center for Lung Biology, University of South Alabama, AL 36688
| | - J R Griswold
- Chemical and Biomolecular Engineering, University of South Alabama, AL 36688
| | - Joshua Deal
- Pharmacology, University of South Alabama, AL 36688
- Center for Lung Biology, University of South Alabama, AL 36688
| | - Naga Annamdevula
- Pharmacology, University of South Alabama, AL 36688
- Center for Lung Biology, University of South Alabama, AL 36688
| | | | - Samuel Mayes
- Chemical and Biomolecular Engineering, University of South Alabama, AL 36688
| | - Craig Browning
- Chemical and Biomolecular Engineering, University of South Alabama, AL 36688
| | - Marina Parker
- Chemical and Biomolecular Engineering, University of South Alabama, AL 36688
| | - Silas J Leavelsey
- Pharmacology, University of South Alabama, AL 36688
- Center for Lung Biology, University of South Alabama, AL 36688
- Chemical and Biomolecular Engineering, University of South Alabama, AL 36688
| |
Collapse
|
31
|
Deal J, Rich TC, Leavesley SJ. Optimizing channel selection for excitation-scanning hyperspectral imaging. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2019; 10881:108811B. [PMID: 34045784 PMCID: PMC8151237 DOI: 10.1117/12.2510784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
A major benefit of fluorescence microscopy is the now plentiful selection of fluorescent markers. These labels can be chosen to serve complementary functions, such as tracking labeled subcellular molecules near demarcated organelles. However, with the standard 3 or 4 emission channels, multiple label detection is restricted to segregated regions of the electromagnetic spectrum, as in RGB coloring. Hyperspectral imaging allows the user to discern many fluorescence labels by their unique spectral properties, provided there is significant differentiation of their emission spectra. The cost of this technique is often an increase in gain or exposure time to accommodate the signal reduction from separating the signal into many discrete excitation or emission channels. Recent advances in hyperspectral imaging have allowed the acquisition of more signal in a shorter time period by scanning the excitation spectra of fluorophores. Here, we explore the selection of optimal channels for both significant signal separation and sufficient signal detection using excitation-scanning hyperspectral imaging. Excitation spectra were obtained using a custom inverted microscope (TE-2000, Nikon Instruments) with a Xe arc lamp and thin film tunable filter array (VersaChrome, Semrock, Inc.) Tunable filters had bandwidths between 13 and 17 nm. Scans utilized excitation wavelengths between 340 nm and 550 nm. Hyperspectral image stacks were generated and analyzed using ENVI and custom MATLAB scripts. Among channel consideration criteria were: number of channels, spectral range of scan, spacing of center wavelengths, and acquisition time.
Collapse
Affiliation(s)
- Joshua Deal
- Department of Chemical & Biomolecular Engineering, University of South Alabama
- Center for Lung Biology, University of South Alabama
- Department of Pharmacology, University of South Alabama
| | - Thomas C Rich
- Center for Lung Biology, University of South Alabama
- Department of Pharmacology, University of South Alabama
| | - Silas J Leavesley
- Department of Chemical & Biomolecular Engineering, University of South Alabama
- Center for Lung Biology, University of South Alabama
- Department of Pharmacology, University of South Alabama
| |
Collapse
|
32
|
Greenwald EC, Mehta S, Zhang J. Genetically Encoded Fluorescent Biosensors Illuminate the Spatiotemporal Regulation of Signaling Networks. Chem Rev 2018; 118:11707-11794. [PMID: 30550275 PMCID: PMC7462118 DOI: 10.1021/acs.chemrev.8b00333] [Citation(s) in RCA: 353] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cellular signaling networks are the foundation which determines the fate and function of cells as they respond to various cues and stimuli. The discovery of fluorescent proteins over 25 years ago enabled the development of a diverse array of genetically encodable fluorescent biosensors that are capable of measuring the spatiotemporal dynamics of signal transduction pathways in live cells. In an effort to encapsulate the breadth over which fluorescent biosensors have expanded, we endeavored to assemble a comprehensive list of published engineered biosensors, and we discuss many of the molecular designs utilized in their development. Then, we review how the high temporal and spatial resolution afforded by fluorescent biosensors has aided our understanding of the spatiotemporal regulation of signaling networks at the cellular and subcellular level. Finally, we highlight some emerging areas of research in both biosensor design and applications that are on the forefront of biosensor development.
Collapse
Affiliation(s)
- Eric C Greenwald
- University of California , San Diego, 9500 Gilman Drive, BRFII , La Jolla , CA 92093-0702 , United States
| | - Sohum Mehta
- University of California , San Diego, 9500 Gilman Drive, BRFII , La Jolla , CA 92093-0702 , United States
| | - Jin Zhang
- University of California , San Diego, 9500 Gilman Drive, BRFII , La Jolla , CA 92093-0702 , United States
| |
Collapse
|
33
|
Hoffman BU, Baba Y, Griffith TN, Mosharov EV, Woo SH, Roybal DD, Karsenty G, Patapoutian A, Sulzer D, Lumpkin EA. Merkel Cells Activate Sensory Neural Pathways through Adrenergic Synapses. Neuron 2018; 100:1401-1413.e6. [PMID: 30415995 DOI: 10.1016/j.neuron.2018.10.034] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 09/21/2018] [Accepted: 10/22/2018] [Indexed: 01/06/2023]
Abstract
Epithelial-neuronal signaling is essential for sensory encoding in touch, itch, and nociception; however, little is known about the release mechanisms and neurotransmitter receptors through which skin cells govern neuronal excitability. Merkel cells are mechanosensory epidermal cells that have long been proposed to activate neuronal afferents through chemical synaptic transmission. We employed a set of classical criteria for chemical neurotransmission as a framework to test this hypothesis. RNA sequencing of adult mouse Merkel cells demonstrated that they express presynaptic molecules and biosynthetic machinery for adrenergic transmission. Moreover, live-cell imaging directly demonstrated that Merkel cells mediate activity- and VMAT-dependent release of fluorescent catecholamine neurotransmitter analogs. Touch-evoked firing in Merkel-cell afferents was inhibited either by pre-synaptic silencing of SNARE-mediated vesicle release from Merkel cells or by neuronal deletion of β2-adrenergic receptors. Together, these results identify both pre- and postsynaptic mechanisms through which Merkel cells excite mechanosensory afferents to encode gentle touch. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Benjamin U Hoffman
- Department of Physiology & Cellular Biophysics, Columbia University, New York, NY, USA; Program in Neurobiology & Behavior, Columbia University, New York, NY, USA
| | - Yoshichika Baba
- Department of Physiology & Cellular Biophysics, Columbia University, New York, NY, USA
| | - Theanne N Griffith
- Department of Physiology & Cellular Biophysics, Columbia University, New York, NY, USA
| | - Eugene V Mosharov
- Departments of Psychiatry, Neurology, and Pharmacology, Columbia University: Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Seung-Hyun Woo
- The Scripps Research Institute & Howard Hughes Medical Institute, La Jolla, CA, USA
| | - Daniel D Roybal
- Pharmacology Graduate Program, Columbia University, New York, NY, USA
| | - Gerard Karsenty
- Department of Genetics and Development, Columbia University, New York, NY, USA
| | - Ardem Patapoutian
- The Scripps Research Institute & Howard Hughes Medical Institute, La Jolla, CA, USA
| | - David Sulzer
- Departments of Psychiatry, Neurology, and Pharmacology, Columbia University: Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Ellen A Lumpkin
- Department of Physiology & Cellular Biophysics, Columbia University, New York, NY, USA; Program in Neurobiology & Behavior, Columbia University, New York, NY, USA; Department of Dermatology, Columbia University, New York, NY, USA.
| |
Collapse
|
34
|
Annamdevula NS, Sweat R, Griswold JR, Trinh K, Hoffman C, West S, Deal J, Britain AL, Jalink K, Rich TC, Leavesley SJ. Spectral imaging of FRET-based sensors reveals sustained cAMP gradients in three spatial dimensions. Cytometry A 2018; 93:1029-1038. [PMID: 30176184 DOI: 10.1002/cyto.a.23572] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 06/21/2018] [Accepted: 07/09/2018] [Indexed: 11/10/2022]
Abstract
Cyclic AMP is a ubiquitous second messenger that orchestrates a variety of cellular functions over different timescales. The mechanisms underlying specificity within this signaling pathway are still not well understood. Several lines of evidence suggest the existence of spatial cAMP gradients within cells, and that compartmentalization underlies specificity within the cAMP signaling pathway. However, to date, no studies have visualized cAMP gradients in three spatial dimensions (3D: x, y, z).This is in part due to the limitations of FRET-based cAMP sensors, specifically the low signal-to-noise ratio intrinsic to all intracellular FRET probes. Here, we overcome this limitation, at least in part, by implementing spectral imaging approaches to estimate FRET efficiency when multiple fluorescent labels are used and when signals are measured from weakly expressed fluorescent proteins in the presence of background autofluorescence and stray light. Analysis of spectral image stacks in two spatial dimensions (2D) from single confocal slices indicates little or no cAMP gradients formed within pulmonary microvascular endothelial cells (PMVECs) under baseline conditions or following 10 min treatment with the adenylyl cyclase activator forskolin. However, analysis of spectral image stacks in 3D demonstrates marked cAMP gradients from the apical to basolateral face of PMVECs. Results demonstrate that spectral imaging approaches can be used to assess cAMP gradients-and in general gradients in fluorescence and FRET-within intact cells. Results also demonstrate that 2D imaging studies of localized fluorescence signals and, in particular, cAMP signals, whether using epifluorescence or confocal microscopy, may lead to erroneous conclusions about the existence and/or magnitude of gradients in either FRET or the underlying cAMP signals. Thus, with the exception of cellular structures that can be considered in one spatial dimension, such as neuronal processes, 3D measurements are required to assess mechanisms underlying compartmentalization and specificity within intracellular signaling pathways.
Collapse
Affiliation(s)
- Naga S Annamdevula
- Department of Chemical & Biomolecular Engineering, University of South Alabama, Mobile, Alabama.,Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Rachel Sweat
- Department of Chemical & Biomolecular Engineering, University of South Alabama, Mobile, Alabama
| | - John R Griswold
- Department of Chemical & Biomolecular Engineering, University of South Alabama, Mobile, Alabama
| | - Kenny Trinh
- Department of Chemical & Biomolecular Engineering, University of South Alabama, Mobile, Alabama
| | - Chase Hoffman
- Medical Sciences, University of South Alabama, Mobile, Alabama
| | - Savannah West
- Department of Biomedical Sciences, University of South Alabama, Mobile, Alabama
| | - Joshua Deal
- Department of Chemical & Biomolecular Engineering, University of South Alabama, Mobile, Alabama.,Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Andrea L Britain
- Center for Lung Biology, University of South Alabama, Mobile, Alabama.,Department of Pharmacology, University of South Alabama, Mobile, Alabama
| | - Kees Jalink
- The Netherlands Cancer Institute and van Leeuwenhoek Center for Advanced Microscopy, Amsterdam, the Netherlands
| | - Thomas C Rich
- Center for Lung Biology, University of South Alabama, Mobile, Alabama.,Department of Pharmacology, University of South Alabama, Mobile, Alabama.,College of Engineering, University of South Alabama, Mobile, Alabama
| | - Silas J Leavesley
- Department of Chemical & Biomolecular Engineering, University of South Alabama, Mobile, Alabama.,Center for Lung Biology, University of South Alabama, Mobile, Alabama.,Department of Pharmacology, University of South Alabama, Mobile, Alabama
| |
Collapse
|
35
|
Filadi R, Basso E, Lefkimmiatis K, Pozzan T. Beyond Intracellular Signaling: The Ins and Outs of Second Messengers Microdomains. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 981:279-322. [PMID: 29594866 DOI: 10.1007/978-3-319-55858-5_12] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A typical characteristic of eukaryotic cells compared to prokaryotes is represented by the spatial heterogeneity of the different structural and functional components: for example, most of the genetic material is surrounded by a highly specific membrane structure (the nuclear membrane), continuous with, yet largely different from, the endoplasmic reticulum (ER); oxidative phosphorylation is carried out by organelles enclosed by a double membrane, the mitochondria; in addition, distinct domains, enriched in specific proteins, are present in the plasma membrane (PM) of most cells. Less obvious, but now generally accepted, is the notion that even the concentration of small molecules such as second messengers (Ca2+ and cAMP in particular) can be highly heterogeneous within cells. In the case of most organelles, the differences in the luminal levels of second messengers depend either on the existence on their membrane of proteins that allow the accumulation/release of the second messenger (e.g., in the case of Ca2+, pumps, exchangers or channels), or on the synthesis and degradation of the specific molecule within the lumen (the autonomous intramitochondrial cAMP system). It needs stressing that the existence of a surrounding membrane does not necessarily imply the existence of a gradient between the cytosol and the organelle lumen. For example, the nuclear membrane is highly permeable to both Ca2+ and cAMP (nuclear pores are permeable to solutes up to 50 kDa) and differences in [Ca2+] or [cAMP] between cytoplasm and nucleoplasm are not seen in steady state and only very transiently during cell activation. A similar situation has been observed, as far as Ca2+ is concerned, in peroxisomes.
Collapse
Affiliation(s)
- Riccardo Filadi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Emy Basso
- Institute of Neuroscience, Padova Section, National Research Council, Padova, Italy
| | - Konstantinos Lefkimmiatis
- Institute of Neuroscience, Padova Section, National Research Council, Padova, Italy
- Venetian Institute of Molecular Medicine, Padova, Italy
| | - Tullio Pozzan
- Department of Biomedical Sciences, University of Padova, Padova, Italy.
- Institute of Neuroscience, Padova Section, National Research Council, Padova, Italy.
- Venetian Institute of Molecular Medicine, Padova, Italy.
| |
Collapse
|
36
|
Spät A, Szanda G. Mitochondrial cAMP and Ca 2+ metabolism in adrenocortical cells. Pflugers Arch 2018; 470:1141-1148. [PMID: 29876637 DOI: 10.1007/s00424-018-2157-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/14/2018] [Accepted: 05/17/2018] [Indexed: 01/22/2023]
Abstract
The biological effects of physiological stimuli of adrenocortical glomerulosa cells are predominantly mediated by the Ca2+ and the cAMP signal transduction pathways. The complex interplay between these signalling systems fine-tunes aldosterone secretion. In addition to the well-known cytosolic interactions, a novel intramitochondrial Ca2+-cAMP interplay has been recently recognised. The cytosolic Ca2+ signal is rapidly transferred into the mitochondrial matrix where it activates Ca2+-sensitive dehydrogenases, thus enhancing the formation of NADPH, a cofactor of steroid synthesis. Quite a few cell types, including H295R adrenocortical cells, express the soluble adenylyl cyclase within the mitochondria and the elevation of mitochondrial [Ca2+] activates the enzyme, thus resulting in the Ca2+-dependent formation of cAMP within the mitochondrial matrix. On the other hand, mitochondrial cAMP (mt-cAMP) potentiates the transfer of cytosolic Ca2+ into the mitochondrial matrix. This cAMP-mediated positive feedback control of mitochondrial Ca2+ uptake may facilitate the rapid hormonal response to emergency situations since knockdown of soluble adenylyl cyclase attenuates aldosterone production whereas overexpression of the enzyme facilitates steroidogenesis in vitro. Moreover, the mitochondrial Ca2+-mt-cAMP-Ca2+ uptake feedback loop is not a unique feature of adrenocortical cells; a similar signalling system has been described in HeLa cells as well.
Collapse
Affiliation(s)
- András Spät
- Department of Physiology, Semmelweis University Medical School, POB 2, Budapest, 1428, Hungary.
- MTA-SE Laboratory of Molecular Physiology, Semmelweis University, Hungarian Academy of Sciences, Budapest, Hungary.
| | - Gergő Szanda
- Department of Physiology, Semmelweis University Medical School, POB 2, Budapest, 1428, Hungary
- MTA-SE Laboratory of Molecular Physiology, Semmelweis University, Hungarian Academy of Sciences, Budapest, Hungary
| |
Collapse
|
37
|
Adenylate Cyclases of Trypanosoma brucei, Environmental Sensors and Controllers of Host Innate Immune Response. Pathogens 2018; 7:pathogens7020048. [PMID: 29693583 PMCID: PMC6027212 DOI: 10.3390/pathogens7020048] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/12/2018] [Accepted: 04/20/2018] [Indexed: 12/12/2022] Open
Abstract
Trypanosoma brucei, etiological agent of Sleeping Sickness in Africa, is the prototype of African trypanosomes, protozoan extracellular flagellate parasites transmitted by saliva (Salivaria). In these parasites the molecular controls of the cell cycle and environmental sensing are elaborate and concentrated at the flagellum. Genomic analyses suggest that these parasites appear to differ considerably from the host in signaling mechanisms, with the exception of receptor-type adenylate cyclases (AC) that are topologically similar to receptor-type guanylate cyclase (GC) of higher eukaryotes but control a new class of cAMP targets of unknown function, the cAMP response proteins (CARPs), rather than the classical protein kinase A cAMP effector (PKA). T. brucei possesses a large polymorphic family of ACs, mainly associated with the flagellar membrane, and these are involved in inhibition of the innate immune response of the host prior to the massive release of immunomodulatory factors at the first peak of parasitemia. Recent evidence suggests that in T. brucei several insect-specific AC isoforms are involved in social motility, whereas only a few AC isoforms are involved in cytokinesis control of bloodstream forms, attesting that a complex signaling pathway is required for environmental sensing. In this review, after a general update on cAMP signaling pathway and the multiple roles of cAMP, I summarize the existing knowledge of the mechanisms by which pathogenic microorganisms modulate cAMP levels to escape immune defense.
Collapse
|
38
|
Agarwal SR, Gratwohl J, Cozad M, Yang PC, Clancy CE, Harvey RD. Compartmentalized cAMP Signaling Associated With Lipid Raft and Non-raft Membrane Domains in Adult Ventricular Myocytes. Front Pharmacol 2018; 9:332. [PMID: 29740315 PMCID: PMC5925456 DOI: 10.3389/fphar.2018.00332] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/21/2018] [Indexed: 11/23/2022] Open
Abstract
Aim: Confining cAMP production to discrete subcellular locations makes it possible for this ubiquitous second messenger to elicit unique functional responses. Yet, factors that determine how and where the production of this diffusible signaling molecule occurs are incompletely understood. The fluid mosaic model originally proposed that signal transduction occurs through random interactions between proteins diffusing freely throughout the plasma membrane. However, it is now known that the movement of membrane proteins is restricted, suggesting that the plasma membrane is segregated into distinct microdomains where different signaling proteins can be concentrated. In this study, we examined what role lipid raft and non-raft membrane domains play in compartmentation of cAMP signaling in adult ventricular myocytes. Methods and Results: The freely diffusible fluorescence resonance energy transfer-based biosensor Epac2-camps was used to measure global cytosolic cAMP responses, while versions of the probe targeted to lipid raft (Epac2-MyrPalm) and non-raft (Epac2-CAAX) domains were used to monitor local cAMP production near the plasma membrane. We found that β-adrenergic receptors, which are expressed in lipid raft and non-raft domains, produce cAMP responses near the plasma membrane that are distinctly different from those produced by E-type prostaglandin receptors, which are expressed exclusively in non-raft domains. We also found that there are differences in basal cAMP levels associated with lipid raft and non-raft domains, and that this can be explained by differences in basal adenylyl cyclase activity associated with each of these membrane environments. In addition, we found evidence that phosphodiesterases 2, 3, and 4 work together in regulating cAMP activity associated with both lipid raft and non-raft domains, while phosphodiesterase 3 plays a more prominent role in the bulk cytoplasmic compartment. Conclusion: These results suggest that different membrane domains contribute to the formation of distinct pools of cAMP under basal conditions as well as following receptor stimulation in adult ventricular myocytes.
Collapse
Affiliation(s)
- Shailesh R Agarwal
- Department of Pharmacology, University of Nevada, Reno, Reno, NV, United States
| | - Jackson Gratwohl
- Department of Pharmacology, University of Nevada, Reno, Reno, NV, United States
| | - Mia Cozad
- Department of Pharmacology, University of Nevada, Reno, Reno, NV, United States
| | - Pei-Chi Yang
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Colleen E Clancy
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Robert D Harvey
- Department of Pharmacology, University of Nevada, Reno, Reno, NV, United States
| |
Collapse
|
39
|
Johnstone TB, Agarwal SR, Harvey RD, Ostrom RS. cAMP Signaling Compartmentation: Adenylyl Cyclases as Anchors of Dynamic Signaling Complexes. Mol Pharmacol 2018; 93:270-276. [PMID: 29217670 PMCID: PMC5820540 DOI: 10.1124/mol.117.110825] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 12/04/2017] [Indexed: 11/22/2022] Open
Abstract
It is widely accepted that cAMP signaling is compartmentalized within cells. However, our knowledge of how receptors, cAMP signaling enzymes, effectors, and other key proteins form specific signaling complexes to regulate specific cell responses is limited. The multicomponent nature of these systems and the spatiotemporal dynamics involved as proteins interact and move within a cell make cAMP responses highly complex. Adenylyl cyclases, the enzymatic source of cAMP production, are key starting points for understanding cAMP compartments and defining the functional signaling complexes. Three basic elements are required to form a signaling compartment. First, a localized signal is generated by a G protein-coupled receptor paired to one or more of the nine different transmembrane adenylyl cyclase isoforms that generate the cAMP signal in the cytosol. The diffusion of cAMP is subsequently limited by several factors, including expression of any number of phosphodiesterases (of which there are 24 genes plus spice variants). Finally, signal response elements are differentially localized to respond to cAMP produced within each locale. A-kinase-anchoring proteins, of which there are 43 different isoforms, facilitate this by targeting protein kinase A to specific substrates. Thousands of potential combinations of these three elements are possible in any given cell type, making the characterization of cAMP signaling compartments daunting. This review will focus on what is known about how cells organize cAMP signaling components as well as identify the unknowns. We make an argument for adenylyl cyclases being central to the formation and maintenance of these signaling complexes.
Collapse
Affiliation(s)
- Timothy B Johnstone
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (T.B.J., R.S.O.); and Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno Nevada (S.R.A., R.D.H.)
| | - Shailesh R Agarwal
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (T.B.J., R.S.O.); and Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno Nevada (S.R.A., R.D.H.)
| | - Robert D Harvey
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (T.B.J., R.S.O.); and Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno Nevada (S.R.A., R.D.H.)
| | - Rennolds S Ostrom
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (T.B.J., R.S.O.); and Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno Nevada (S.R.A., R.D.H.)
| |
Collapse
|
40
|
Mathematical Modelling of Nitric Oxide/Cyclic GMP/Cyclic AMP Signalling in Platelets. Int J Mol Sci 2018; 19:ijms19020612. [PMID: 29462984 PMCID: PMC5855834 DOI: 10.3390/ijms19020612] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 02/04/2023] Open
Abstract
Platelet activation contributes to normal haemostasis but also to pathologic conditions like stroke and cardiac infarction. Signalling by cGMP and cAMP inhibit platelet activation and are therefore attractive targets for thrombosis prevention. However, extensive cross-talk between the cGMP and cAMP signalling pathways in multiple tissues complicates the selective targeting of their activities. We have used mathematical modelling based on experimental data from the literature to quantify the steady state behaviour of nitric oxide (NO)/cGMP/cAMP signalling in platelets. The analysis provides an assessment of NO-induced cGMP synthesis and PKG activation as well as cGMP-mediated cAMP and PKA activation though modulation of phosphodiesterase (PDE2 and 3) activities. Both one- and two-compartment models of platelet cyclic nucleotide signalling are presented. The models provide new insight for understanding how NO signalling to cGMP and indirectly cAMP, can inhibit platelet shape-change, the initial step of platelet activation. Only the two-compartment models could account for the experimental observation that NO-mediated PKA activation can occur when the bulk platelet cAMP level is unchanged. The models revealed also a potential for hierarchical interplay between the different platelet phosphodiesterases. Specifically, the models predict, unexpectedly, a strong effect of pharmacological inhibitors of cGMP-specific PDE5 on the cGMP/cAMP cross-talk. This may explain the successful use of weak PDE5-inhibitors, such as dipyridamole, in anti-platelet therapy. In conclusion, increased NO signalling or PDE5 inhibition are attractive ways of increasing cGMP-cAMP cross-talk selectively in platelets.
Collapse
|
41
|
Pavlaki N, Nikolaev VO. Imaging of PDE2- and PDE3-Mediated cGMP-to-cAMP Cross-Talk in Cardiomyocytes. J Cardiovasc Dev Dis 2018; 5:jcdd5010004. [PMID: 29367582 PMCID: PMC5872352 DOI: 10.3390/jcdd5010004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 01/16/2018] [Accepted: 01/17/2018] [Indexed: 12/13/2022] Open
Abstract
Cyclic nucleotides 3′,5′-cyclic adenosine monophosphate (cAMP) and 3′,5′-cyclic guanosine monophosphate (cGMP) are important second messengers that regulate cardiovascular function and disease by acting in discrete subcellular microdomains. Signaling compartmentation at these locations is often regulated by phosphodiesterases (PDEs). Some PDEs are also involved in the cross-talk between the two second messengers. The purpose of this review is to summarize and highlight recent findings about the role of PDE2 and PDE3 in cardiomyocyte cyclic nucleotide compartmentation and visualization of this process using live cell imaging techniques.
Collapse
Affiliation(s)
- Nikoleta Pavlaki
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany.
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany.
| |
Collapse
|
42
|
Abstract
Fertilization is exceptionally complex and, depending on the species, happens in entirely different environments. External fertilizers in aquatic habitats, like marine invertebrates or fish, release their gametes into the seawater or freshwater, whereas sperm from most internal fertilizers like mammals cross the female genital tract to make their way to the egg. Various chemical and physical cues guide sperm to the egg. Quite generally, these cues enable signaling pathways that ultimately evoke a cellular Ca2+ response that modulates the waveform of the flagellar beat and, hence, the swimming path. To cope with the panoply of challenges to reach and fertilize the egg, sperm from different species have developed their own unique repertoire of signaling molecules and mechanisms. Here, we review the differences and commonalities for sperm sensory signaling in marine invertebrates (sea urchin), fish (zebrafish), and mammals (mouse, human).
Collapse
Affiliation(s)
- Dagmar Wachten
- Minerva Max Planck Research Group, Molecular Physiology, Center of Advanced European Studies and Research (caesar), 53175 Bonn, Germany
| | - Jan F Jikeli
- Minerva Max Planck Research Group, Molecular Physiology, Center of Advanced European Studies and Research (caesar), 53175 Bonn, Germany
| | - U Benjamin Kaupp
- Department Molecular Sensory Systems, Center of Advanced European Studies and Research (caesar), 53175 Bonn, Germany
| |
Collapse
|
43
|
Rossi ML, Rubbini G, Martini M, Canella R, Fesce R. Forskolin and protein kinase inhibitors differentially affect hair cell potassium currents and transmitter release at the cytoneural junction in the isolated frog labyrinth. Neuroscience 2017; 357:20-36. [PMID: 28576732 DOI: 10.1016/j.neuroscience.2017.05.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 05/20/2017] [Accepted: 05/23/2017] [Indexed: 11/19/2022]
Abstract
The post-transductional elaboration of sensory input at the frog semicircular canal has been studied by correlating the effects of drugs that interfere with phosphorylation processes on: (i) potassium conductances in isolated hair cell and (ii) transmitter release at the cytoneural junction in the intact labyrinth. At hair cells, delayed potassium currents (IKD) undergo voltage- and time-dependent inactivation; inactivation removal requires ATP, is sensitive to kinase blockade, but is unaffected by exogenous application of cyclic nucleotides. We report here that forskolin, an activator of endogenous adenylyl cyclase, enhances IKD inactivation removal in isolated hair cells, but produces an overall decrease in IKD amplitude consistent with the direct blocking action of the drug on several families of K channels. In the intact labyrinth, forskolin enhances transmitter release, consistent with such depression of K conductances. Kinase blockers - H-89 and KT5823 - have been shown to reduce IKD inactivation removal and IKD amplitude at isolated hair cells. In the labyrinth, the effects of these drugs on junctional activity are quite variable, with predominant inhibition of transmitter release, rather than the enhancement expected from the impairment of K currents. The overall action of forskolin and kinase inhibitors on K conductances is similar (depression), but they have opposite effects on transmitter release: this indicates that some intermediate steps between the bioelectric control of hair cell membrane potential and transmitter release are affected in opposite ways and therefore are presumably regulated by protein phosphorylation.
Collapse
Affiliation(s)
- Maria Lisa Rossi
- Dipartimento di Scienze della Vita e Biotecnologie, Ferrara University, Ferrara, Italy.
| | - Gemma Rubbini
- Dipartimento di Scienze della Vita e Biotecnologie, Ferrara University, Ferrara, Italy
| | - Marta Martini
- Dipartimento di Scienze della Vita e Biotecnologie, Ferrara University, Ferrara, Italy
| | - Rita Canella
- Dipartimento di Scienze della Vita e Biotecnologie, Ferrara University, Ferrara, Italy
| | - Riccardo Fesce
- Centre of Neuroscience, DISTA, Insubria University, Varese, Italy
| |
Collapse
|
44
|
Rozier K, Bondarenko VE. Distinct physiological effects of β1- and β2-adrenoceptors in mouse ventricular myocytes: insights from a compartmentalized mathematical model. Am J Physiol Cell Physiol 2017; 312:C595-C623. [DOI: 10.1152/ajpcell.00273.2016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 01/03/2017] [Accepted: 01/18/2017] [Indexed: 01/08/2023]
Abstract
The β1- and β2-adrenergic signaling systems play different roles in the functioning of cardiac cells. Experimental data show that the activation of the β1-adrenergic signaling system produces significant inotropic, lusitropic, and chronotropic effects in the heart, whereas the effects of the β2-adrenergic signaling system is less apparent. In this paper, a comprehensive compartmentalized experimentally based mathematical model of the combined β1- and β2-adrenergic signaling systems in mouse ventricular myocytes is developed to simulate the experimental findings and make testable predictions of the behavior of the cardiac cells under different physiological conditions. Simulations describe the dynamics of major signaling molecules in different subcellular compartments; kinetics and magnitudes of phosphorylation of ion channels, transporters, and Ca2+ handling proteins; modifications of action potential shape and duration; and [Ca2+]i and [Na+]i dynamics upon stimulation of β1- and β2-adrenergic receptors (β1- and β2-ARs). The model reveals physiological conditions when β2-ARs do not produce significant physiological effects and when their effects can be measured experimentally. Simulations demonstrated that stimulation of β2-ARs with isoproterenol caused a marked increase in the magnitude of the L-type Ca2+ current, [Ca2+]i transient, and phosphorylation of phospholamban only upon additional application of pertussis toxin or inhibition of phosphodiesterases of type 3 and 4. The model also made testable predictions of the changes in magnitudes of [Ca2+]i and [Na+]i fluxes, the rate of decay of [Na+]i concentration upon both combined and separate stimulation of β1- and β2-ARs, and the contribution of phosphorylation of PKA targets to the changes in the action potential and [Ca2+]i transient.
Collapse
Affiliation(s)
- Kelvin Rozier
- Department of Mathematics and Statistics, Georgia State University, Atlanta, Georgia; and
| | - Vladimir E. Bondarenko
- Department of Mathematics and Statistics, Georgia State University, Atlanta, Georgia; and
- Neuroscience Institute, Georgia State University, Atlanta, Georgia
| |
Collapse
|
45
|
Torres-Quesada O, Röck R, Stefan E. Systematic Quantification of GPCR/cAMP-Controlled Protein Kinase A Interactions. Horm Metab Res 2017; 49:240-249. [PMID: 28427097 DOI: 10.1055/s-0042-110791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The diffusible second messenger cyclic AMP (cAMP) originates from multiple G protein-coupled receptor (GPCR) cascades activating the intracellular key effector protein kinase A (PKA). Spatially and temporally restricted cAMP-fluxes are directly sensed by macromolecular PKA complexes. The consequences are alterations of molecular interactions, which lead to activation of compartmentalized PKA phosphotransferase activities, regulating a vast array of cellular functions. To decode cell-type and cell-compartment specific PKA functions, the spatio-temporal dynamics of small molecule:protein interactions, protein:protein interactions (PPIs), cAMP-mobilization, and phosphotransferase activities need to be determined directly in the appropriate cellular context. A collection of cell-based reporters has been developed to either visualize or quantitatively measure kinase activities or PKA complex formation/dissociation. In this review, we list a collection of unimolecular and bimolecular PKA biosensors, followed by the specification of the modular design of a Renilla luciferase based protein-fragment complementation assay (PCA) platform for measuring PKA network interactions. We discuss the application spectrum of the PCA reporter to identify, quantify, and dissect dynamic and transient PKA complexes downstream of specific GPCR activities. We specify the implementation of a PCA PKA platform to systematically quantify the concurrent involvement of receptor-cAMP signaling, post-translational modifications, and kinase subunit mutations/perturbations in PKA activation. The systematic quantification of transient PKA network interactions will contribute to a better understanding how GPCR-recognized input signals are streamlined through the compartmentalized and cAMP-interacting PKA signalosome.
Collapse
Affiliation(s)
- O Torres-Quesada
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - R Röck
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - E Stefan
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
46
|
Sharma S, Visweswariah SS. Illuminating Cyclic Nucleotides: Sensors for cAMP and cGMP and Their Application in Live Cell Imaging. J Indian Inst Sci 2017. [DOI: 10.1007/s41745-016-0014-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
47
|
Takeda Y. [Theoretical Investigations into the Quantitative Mechanisms Underlying the Regulation of [cAMP]i, Membrane Excitability and [Ca(2+)]i during GLP-1 Stimulation in Pancreatic β Cells]. YAKUGAKU ZASSHI 2017; 136:467-71. [PMID: 26935088 DOI: 10.1248/yakushi.15-00246-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Upon elevation of plasma glucose concentration, pancreatic β-cells generate bursts of action potentials to induce cyclic changes in [Ca(2+)]i regulating insulin release. Glucose-dependent insulin secretion is synergistically enhanced by glucagon-like peptide-1 (GLP-1), which increases [cAMP]i and activates protein kinase A (PKA) and exchange protein activated by cAMP (Epac). The insulinotropic effect of GLP-1 is mediated, at least in part, by modulating multiple ion channels/transporters at the plasma membrane and ER through PKA- and EPAC-dependent mechanisms, which increase membrane excitability and intracellular Ca(2+) release. However, because of complex interactions between multiple cellular factors involved in the GLP-1 effects, quantitative aspects of the molecular/ionic mechanisms have not yet been determined. We thus performed simulation studies and mathematical analysis to investigate how GLP-1 signals control [cAMP]i and subsequently modify the bursting activities and Ca(2+) dynamics. First, a GLP-1 receptor signal transduction model was developed and introduced to our β-cells model. Secondly, modulatory effects of PKA/Epac on ion channels/transporters were incorporated based on experimental studies. Increases in the frequency and duration of the bursting activity observed during GLP-1 stimulation were well reconstructed by our model, and lead potential analysis quantitatively determined the functional role of each ion channel/transporter in modifying the burst pattern. Finally, an IP3R model was developed to reproduce GLP-1-induced Ca(2+) transients/oscillations. Instantaneous equilibrium analysis and bifurcation analysis also elucidated the quantitative mechanisms involved in generating IP3R-mediated Ca(2+) mobilization. The results of this theoretical analysis of the effects of GLP-1 on membrane excitability/Ca(2+) dynamics are discussed in this review.
Collapse
Affiliation(s)
- Yukari Takeda
- Department of Bioinformatics, College of Life Sciences, Ritsumeikan University
| |
Collapse
|
48
|
Rich TC, Annamdevula N, Trinh K, Britain AL, Mayes SA, Griswold JR, Deal J, Hoffman C, West S, Leavesley SJ. 5D imaging approaches reveal the formation of distinct intracellular cAMP spatial gradients. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2017; 10070:100700R. [PMID: 34054188 PMCID: PMC8157067 DOI: 10.1117/12.2253164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Cyclic AMP (cAMP) is a ubiquitous second messenger known to differentially regulate many cellular functions. Several lines of evidence suggest that the distribution of cAMP within cells is not uniform. However, to date, no studies have measured the kinetics of 3D cAMP distributions within cells. This is largely due to the low signal-to-noise ratio of FRET-based probes. We previously reported that hyperspectral imaging improves the signal-to-noise ratio of FRET measurements. Here we utilized hyperspectral imaging approaches to measure FRET signals in five dimensions (5D) - three spatial (x, y, z), wavelength (λ), and time (t) - allowing us to visualize cAMP gradients in pulmonary endothelial cells. cAMP levels were measured using a FRET-based sensor (H188) comprised of a cAMP binding domain sandwiched between FRET donor and acceptor - Turquoise and Venus fluorescent proteins. We observed cAMP gradients in response to 0.1 or 1 μM isoproterenol, 0.1 or 1 μM PGE1, or 50 μM forskolin. Forskolin- and isoproterenol-induced cAMP gradients formed from the apical (high cAMP) to basolateral (low cAMP) face of cells. In contrast, PGE1-induced cAMP gradients originated from both the basolateral and apical faces of cells. Data suggest that 2D (x,y) studies of cAMP compartmentalization may lead to erroneous conclusions about the existence of cAMP gradients, and that 3D (x,y,z) studies are required to assess mechanisms of signaling specificity. Results demonstrate that 5D imaging technologies are powerful tools for measuring biochemical processes in discrete subcellular domains. This work was supported by NIH P01HL066299, R01HL058506, S10RR027535, AHA 16PRE27130004 and the Abraham Mitchell Cancer Research Fund.
Collapse
Affiliation(s)
- Thomas C Rich
- Pharmacology, University of South Alabama, AL 36688
- Center for Lung Biology, University of South Alabama, AL 36688
- Basic Medical Sciences Graduate Program, University of South Alabama, AL 36688
| | - Naga Annamdevula
- Center for Lung Biology, University of South Alabama, AL 36688
- Basic Medical Sciences Graduate Program, University of South Alabama, AL 36688
| | - Kenny Trinh
- Chemical and Biomolecular Engineering, University of South Alabama, AL 36688
| | - Andrea L Britain
- Pharmacology, University of South Alabama, AL 36688
- Center for Lung Biology, University of South Alabama, AL 36688
| | - Samuel A Mayes
- Chemical and Biomolecular Engineering, University of South Alabama, AL 36688
| | - John R Griswold
- Chemical and Biomolecular Engineering, University of South Alabama, AL 36688
| | - Joshua Deal
- Center for Lung Biology, University of South Alabama, AL 36688
- Basic Medical Sciences Graduate Program, University of South Alabama, AL 36688
| | | | - Savannah West
- Biomedical Sciences, University of South Alabama, AL 36688
| | - Silas J Leavesley
- Pharmacology, University of South Alabama, AL 36688
- Center for Lung Biology, University of South Alabama, AL 36688
- Basic Medical Sciences Graduate Program, University of South Alabama, AL 36688
- Chemical and Biomolecular Engineering, University of South Alabama, AL 36688
| |
Collapse
|
49
|
Pendin D, Greotti E, Lefkimmiatis K, Pozzan T. Exploring cells with targeted biosensors. J Gen Physiol 2016; 149:1-36. [PMID: 28028123 PMCID: PMC5217087 DOI: 10.1085/jgp.201611654] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 10/26/2016] [Accepted: 12/01/2016] [Indexed: 01/10/2023] Open
Abstract
Cellular signaling networks are composed of multiple pathways, often interconnected, that form complex networks with great potential for cross-talk. Signal decoding depends on the nature of the message as well as its amplitude, temporal pattern, and spatial distribution. In addition, the existence of membrane-bound organelles, which are both targets and generators of messages, add further complexity to the system. The availability of sensors that can localize to specific compartments in live cells and monitor their targets with high spatial and temporal resolution is thus crucial for a better understanding of cell pathophysiology. For this reason, over the last four decades, a variety of strategies have been developed, not only to generate novel and more sensitive probes for ions, metabolites, and enzymatic activity, but also to selectively deliver these sensors to specific intracellular compartments. In this review, we summarize the principles that have been used to target organic or protein sensors to different cellular compartments and their application to cellular signaling.
Collapse
Affiliation(s)
- Diana Pendin
- Neuroscience Institute, National Research Council, Padua Section, 35121 Padua, Italy.,Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy
| | - Elisa Greotti
- Neuroscience Institute, National Research Council, Padua Section, 35121 Padua, Italy.,Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy
| | - Konstantinos Lefkimmiatis
- Neuroscience Institute, National Research Council, Padua Section, 35121 Padua, Italy.,Venetian Institute of Molecular Medicine, 35129 Padua, Italy
| | - Tullio Pozzan
- Neuroscience Institute, National Research Council, Padua Section, 35121 Padua, Italy.,Venetian Institute of Molecular Medicine, 35129 Padua, Italy.,Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy
| |
Collapse
|
50
|
Wilson CM, Roa JN, Cox GK, Tresguerres M, Farrell AP. Introducing a novel mechanism to control heart rate in the ancestral Pacific hagfish. ACTA ACUST UNITED AC 2016; 219:3227-3236. [PMID: 27510962 DOI: 10.1242/jeb.138198] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 08/02/2016] [Indexed: 12/15/2022]
Abstract
Although neural modulation of heart rate is well established among chordate animals, the Pacific hagfish (Eptatretus stoutii) lacks any cardiac innervation, yet it can increase its heart rate from the steady, depressed heart rate seen in prolonged anoxia to almost double its normal normoxic heart rate, an almost fourfold overall change during the 1-h recovery from anoxia. The present study sought mechanistic explanations for these regulatory changes in heart rate. We provide evidence for a bicarbonate-activated, soluble adenylyl cyclase (sAC)-dependent mechanism to control heart rate, a mechanism never previously implicated in chordate cardiac control.
Collapse
Affiliation(s)
- Christopher M Wilson
- Department of Zoology, University of British Columbia, 6270 University Boulevard, Vancouver, British Columbia, Canada V6T 1Z4
| | - Jinae N Roa
- Marine Biology Research Division, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Georgina K Cox
- Department of Zoology, University of British Columbia, 6270 University Boulevard, Vancouver, British Columbia, Canada V6T 1Z4
| | - Martin Tresguerres
- Marine Biology Research Division, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Anthony P Farrell
- Department of Zoology, University of British Columbia, 6270 University Boulevard, Vancouver, British Columbia, Canada V6T 1Z4.,Faculty of Land and Food Systems, University of British Columbia, 2357 Main Mall, Vancouver, British Columbia, Canada V6T 1Z4
| |
Collapse
|