1
|
Ulrich H, Glaser T, Thomas AP. Purinergic signaling in liver disease: calcium signaling and induction of inflammation. Purinergic Signal 2024:10.1007/s11302-024-10044-9. [PMID: 39320433 DOI: 10.1007/s11302-024-10044-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 08/15/2024] [Indexed: 09/26/2024] Open
Abstract
Purinergic signaling regulates many metabolic functions and is implicated in liver physiology and pathophysiology. Liver functionality is modulated by ionotropic P2X and metabotropic P2Y receptors, specifically P2Y1, P2Y2, and P2Y6 subtypes, which physiologically exert their influence through calcium signaling, a key second messenger controlling glucose and fat metabolism in hepatocytes. Purinergic receptors, acting through calcium signaling, play an important role in a range of liver diseases. Ionotropic P2X receptors, such as the P2X7 subtype, and certain metabotropic P2Y receptors can induce aberrant intracellular calcium transients that impact normal hepatocyte function and initiate the activation of other liver cell types, including Kupffer and stellate cells. These P2Y- and P2X-dependent intracellular calcium increases are particularly relevant in hepatic disease states, where stellate and Kupffer cells respond with innate immune reactions to challenges, such as excess fat accumulation, chronic alcohol abuse, or infections, and can eventually lead to liver fibrosis. This review explores the consequences of excessive extracellular ATP accumulation, triggering calcium influx through P2X4 and P2X7 receptors, inflammasome activation, and programmed cell death. In addition, P2Y2 receptors contribute to hepatic steatosis and insulin resistance, while inhibiting the expression of P2Y6 receptors can alleviate alcoholic liver steatosis. Adenosine receptors may also contribute to fibrosis through extracellular matrix production by fibroblasts. Thus, pharmacological modulation of P1 and P2 receptors and downstream calcium signaling may open novel therapeutic avenues.
Collapse
Affiliation(s)
- Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, 05508-000, Brazil.
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA.
| | - Talita Glaser
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, 05508-000, Brazil.
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA.
| | - Andrew P Thomas
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| |
Collapse
|
2
|
Mesa D, Barbieri E, Raimondi A, Freddi S, Miloro G, Jendrisek G, Caldieri G, Quarto M, Schiano Lomoriello I, Malabarba MG, Bresci A, Manetti F, Vernuccio F, Abdo H, Scita G, Lanzetti L, Polli D, Tacchetti C, Pinton P, Bonora M, Di Fiore PP, Sigismund S. A tripartite organelle platform links growth factor receptor signaling to mitochondrial metabolism. Nat Commun 2024; 15:5119. [PMID: 38879572 PMCID: PMC11180189 DOI: 10.1038/s41467-024-49543-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 06/08/2024] [Indexed: 06/19/2024] Open
Abstract
One open question in the biology of growth factor receptors is how a quantitative input (i.e., ligand concentration) is decoded by the cell to produce specific response(s). Here, we show that an EGFR endocytic mechanism, non-clathrin endocytosis (NCE), which is activated only at high ligand concentrations and targets receptor to degradation, requires a tripartite organelle platform involving the plasma membrane (PM), endoplasmic reticulum (ER) and mitochondria. At these contact sites, EGFR-dependent, ER-generated Ca2+ oscillations are sensed by mitochondria, leading to increased metabolism and ATP production. Locally released ATP is required for cortical actin remodeling and EGFR-NCE vesicle fission. The same biochemical circuitry is also needed for an effector function of EGFR, i.e., collective motility. The multiorganelle signaling platform herein described mediates direct communication between EGFR signaling and mitochondrial metabolism, and is predicted to have a broad impact on cell physiology as it is activated by another growth factor receptor, HGFR/MET.
Collapse
Affiliation(s)
- Deborah Mesa
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Andrea Raimondi
- Experimental Imaging Centre, IRCCS San Raffaele Hospital Scientific Institute, Milan, Italy
- Università della Svizzera italiana (USI), Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Stefano Freddi
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Gorana Jendrisek
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Micaela Quarto
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Irene Schiano Lomoriello
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Maria Grazia Malabarba
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Arianna Bresci
- Department of Physics, Politecnico di Milano, Milan, Italy
| | | | | | - Hind Abdo
- IFOM, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Giorgio Scita
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
- IFOM, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Letizia Lanzetti
- Department of Oncology, University of Torino Medical School, Candiolo, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Dario Polli
- Department of Physics, Politecnico di Milano, Milan, Italy
- CNR Institute for Photonics and Nanotechnology (CNR-IFN), Milan, Italy
| | - Carlo Tacchetti
- Experimental Imaging Centre, IRCCS San Raffaele Hospital Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Paolo Pinton
- Department of Medical Sciences, Section of Experimental Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Massimo Bonora
- Department of Medical Sciences, Section of Experimental Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Pier Paolo Di Fiore
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy.
- IEO, European Institute of Oncology IRCCS, Milan, Italy.
| | - Sara Sigismund
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy.
- IEO, European Institute of Oncology IRCCS, Milan, Italy.
| |
Collapse
|
3
|
Coccarelli A, Pant S. On the Ca 2+ elevation in vascular endothelial cells due to inositol trisphosphate-sensitive store receptors activation: A data-driven modeling approach. Comput Biol Med 2023; 164:107111. [PMID: 37540925 DOI: 10.1016/j.compbiomed.2023.107111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 05/18/2023] [Accepted: 05/30/2023] [Indexed: 08/06/2023]
Abstract
Agonist-induced Ca2+ signaling is essential for the regulation of many vital functions in endothelial cells (ECs). A broad range of stimuli elevate the cytosolic Ca2+ concentration by promoting a pathway mediated by inositol 1,4,5 trisphosphate (IP3) which causes Ca2+ release from intracellular stores. Despite its importance, there are very few studies focusing on the quantification of such dynamics in the vascular endothelium. Here, by using data from isolated ECs, we established a minimalistic modeling framework able to quantitatively capture the main features (averaged over a cell population) of the cytosolic Ca2+ response to different IP3 stimulation levels. A suitable description of Ca2+-regulatory function of inositol 1,4,5 trisphosphate receptors (IP3Rs) and corresponding parameter space are identified by comparing the different model variants against experimental mean population data. The same approach is used to numerically assess the relevance of cytosolic Ca2+ buffering, as well as Ca2+ store IP3-sensitivity in the overall cell dynamics. The variability in the dynamics' features observed across the population can be explained (at least in part) through variation of certain model parameters (such as buffering capacity or Ca2+ store sensitivity to IP3). The results, in terms of experimental fitting and validation, support the proposed minimalistic model as a reference framework for the quantification of the EC Ca2+ dynamics induced by IP3Rs activation.
Collapse
Affiliation(s)
- Alberto Coccarelli
- Zienkiewicz Centre for Computational Engineering, Faculty of Science and Engineering, Swansea University, UK.
| | - Sanjay Pant
- Zienkiewicz Centre for Computational Engineering, Faculty of Science and Engineering, Swansea University, UK
| |
Collapse
|
4
|
Besio R, Contento BM, Garibaldi N, Filibian M, Sonntag S, Shmerling D, Tonelli F, Biggiogera M, Brini M, Salmaso A, Jovanovic M, Marini JC, Rossi A, Forlino A. CaMKII inhibition due to TRIC-B loss-of-function dysregulates SMAD signaling in osteogenesis imperfecta. Matrix Biol 2023; 120:43-59. [PMID: 37178987 PMCID: PMC11123566 DOI: 10.1016/j.matbio.2023.05.002] [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: 03/05/2023] [Revised: 05/02/2023] [Accepted: 05/10/2023] [Indexed: 05/15/2023]
Abstract
Ca2+ is a second messenger that regulates a variety of cellular responses in bone, including osteoblast differentiation. Mutations in trimeric intracellular cation channel B (TRIC-B), an endoplasmic reticulum channel specific for K+, a counter ion for Ca2+flux, affect bone and cause a recessive form of osteogenesis imperfecta (OI) with a still puzzling mechanism. Using a conditional Tmem38b knock out mouse, we demonstrated that lack of TRIC-B in osteoblasts strongly impairs skeleton growth and structure, leading to bone fractures. At the cellular level, delayed osteoblast differentiation and decreased collagen synthesis were found consequent to the Ca2+ imbalance and associated with reduced collagen incorporation in the extracellular matrix and poor mineralization. The impaired SMAD signaling detected in mutant mice, and validated in OI patient osteoblasts, explained the osteoblast malfunction. The reduced SMAD phosphorylation and nuclear translocation were mainly caused by alteration in Ca2+ calmodulin kinase II (CaMKII)-mediated signaling and to a less extend by a lower TGF-β reservoir. SMAD signaling, osteoblast differentiation and matrix mineralization were only partially rescued by TGF-β treatment, strengthening the impact of CaMKII-SMAD axes on osteoblast function. Our data established the TRIC-B role in osteoblasts and deepened the contribution of the CaMKII-SMAD signaling in bone.
Collapse
Affiliation(s)
- Roberta Besio
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Barbara M Contento
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Nadia Garibaldi
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Marta Filibian
- Centro Grandi Strumenti, University of Pavia, Pavia, Italy; INFN, Istituto Nazionale di Fisica Nucleare-Pavia Unit, Pavia, Italy
| | - Stephan Sonntag
- PolyGene AG, Rümlang, Switzerland; LIMES-Institute, University of Bonn, Bonn , Germany
| | | | - Francesca Tonelli
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Marco Biggiogera
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Marisa Brini
- Department of Biology, University of Padova, Padova, Italy; Centro Studi per la Neurodegenerazione (CESNE), University of Padova, Padova, Italy
| | - Andrea Salmaso
- Department of Biology, University of Padova, Padova, Italy
| | - Milena Jovanovic
- Section on Heritable Disorders of Bone and Extracellular Matrix, Eunice Kennedy Shiver National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, United States of America
| | - Joan C Marini
- Section on Heritable Disorders of Bone and Extracellular Matrix, Eunice Kennedy Shiver National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, United States of America
| | - Antonio Rossi
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Antonella Forlino
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy.
| |
Collapse
|
5
|
Sarma U, Maiti M, Nair A, Bhadange S, Bansode Y, Srivastava A, Saha B, Mukherjee D. Regulation of STAT3 signaling in IFNγ and IL10 pathways and in their cross-talk. Cytokine 2021; 148:155665. [PMID: 34366205 DOI: 10.1016/j.cyto.2021.155665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 11/17/2022]
Abstract
The pro-inflammatory IFNγ-STAT1 pathway and anti-inflammatory IL10-STAT3 pathway elicit cellular responses primarily utilizing their canonical STATs. However IL10 mediated STAT1 and IFNγ mediated STAT3 activation is also observed, suggesting crosstalk of these functionally opposing signaling pathways can potentially reshape the canonical dynamics both STATs and alter the expression of their target genes. Herein, we measured the dynamics of STATs in response to different doses of IL10 or IFNγ and in their co-stimulation and employed quantitative modeling to understand the regulatory mechanisms controlling signal responses in individual and co-simulation scenarios. Our experiments show, STAT3 in particular, exhibits a bell-shaped dose-response while treated with IFNγ or IL10 and our model quantiatively captured the dose-dependent dynamics of both the STATs in both pathways. The model next predicted and subsequent experiments validated that STAT3 dynamics would robustly remain IL10 specific when subjected to a co-stimulation of both IFNγ and IL10. Genes common to both pathways also exhibited IL10 specific expression during the co-stimulation. The findings thus uncover anovel feature of the IL10-STAT3 signaling axis during pathway crosstalk. Finally, parameter sampling coupled to information theory based analysis showed that bell-shaped signal-response of STAT3 in both pathways is primarily dependent on receptor concentration whereas robustness of IL10-STAT3 signaling axis in co-stimulation results from the negative regulation of the IFNγ pathway.
Collapse
Affiliation(s)
- U Sarma
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India.
| | - M Maiti
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India
| | - A Nair
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India
| | - S Bhadange
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India
| | - Y Bansode
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India
| | - A Srivastava
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India
| | - B Saha
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India
| | - D Mukherjee
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, India.
| |
Collapse
|
6
|
Receptor-specific Ca 2+ oscillation patterns mediated by differential regulation of P2Y purinergic receptors in rat hepatocytes. iScience 2021; 24:103139. [PMID: 34646983 PMCID: PMC8496176 DOI: 10.1016/j.isci.2021.103139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/26/2021] [Accepted: 09/14/2021] [Indexed: 11/21/2022] Open
Abstract
Extracellular agonists linked to inositol-1,4,5-trisphosphate (IP3) formation elicit cytosolic Ca2+ oscillations in many cell types, but despite a common signaling pathway, distinct agonist-specific Ca2+ spike patterns are observed. Using qPCR, we show that rat hepatocytes express multiple purinergic P2Y and P2X receptors (R). ADP acting through P2Y1R elicits narrow Ca2+ oscillations, whereas UTP acting through P2Y2R elicits broad Ca2+ oscillations, with composite patterns observed for ATP. P2XRs do not play a role at physiological agonist levels. The discrete Ca2+ signatures reflect differential effects of protein kinase C (PKC), which selectively modifies the falling phase of the Ca2+ spikes. Negative feedback by PKC limits the duration of P2Y1R-induced Ca2+ spikes in a manner that requires extracellular Ca2+. By contrast, P2Y2R is resistant to PKC negative feedback. Thus, the PKC leg of the bifurcated IP3 signaling pathway shapes unique Ca2+ oscillation patterns that allows for distinct cellular responses to different agonists. Distinct stereotypic Ca2+ oscillations are elicited by P2Y1 and P2Y2 receptors P2X receptors do not contribute to the generation of Ca2+ oscillations Agonist-specific Ca2+ spike shapes reflect discrete modes of PKC negative feedback Bifurcation of IP3/PKC signaling yields unique Ca2+ oscillation signatures
Collapse
|
7
|
Shipman JG, Onyenwoke RU, Sivaraman V. Calcium-Dependent Pulmonary Inflammation and Pharmacological Interventions and Mediators. BIOLOGY 2021; 10:1053. [PMID: 34681152 PMCID: PMC8533358 DOI: 10.3390/biology10101053] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/11/2021] [Accepted: 10/14/2021] [Indexed: 12/17/2022]
Abstract
Pulmonary diseases present a significant burden worldwide and lead to severe morbidity and mortality. Lung inflammation caused by interactions with either viruses, bacteria or fungi is a prominent characteristic of many pulmonary diseases. Tobacco smoke and E-cig use ("vaping") are considered major risk factors in the development of pulmonary disease as well as worsening disease prognosis. However, at present, relatively little is known about the mechanistic actions by which smoking and vaping may worsen the disease. One theory suggests that long-term vaping leads to Ca2+ signaling dysregulation. Ca2+ is an important secondary messenger in signal transduction. Cellular Ca2+ concentrations are mediated by a complex series of pumps, channels, transporters and exchangers that are responsible for triggering various intracellular processes such as cell death, proliferation and secretion. In this review, we provide a detailed understating of the complex series of components that mediate Ca2+ signaling and how their dysfunction may result in pulmonary disease. Furthermore, we summarize the recent literature investigating the negative effects of smoking and vaping on pulmonary disease, cell toxicity and Ca2+ signaling. Finally, we summarize Ca2+-mediated pharmacological interventions that could potentially lead to novel treatments for pulmonary diseases.
Collapse
Affiliation(s)
- Jeffrey G. Shipman
- Department of Biological and Biomedical Sciences, College of Health and Sciences, North Carolina Central University, Durham, NC 27707, USA;
| | - Rob U. Onyenwoke
- Department of Pharmaceutical Sciences, North Carolina Central University, Durham, NC 27707, USA;
- Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA
| | - Vijay Sivaraman
- Department of Biological and Biomedical Sciences, College of Health and Sciences, North Carolina Central University, Durham, NC 27707, USA;
| |
Collapse
|
8
|
Knocking out TMEM38B in human foetal osteoblasts hFOB 1.19 by CRISPR/Cas9: A model for recessive OI type XIV. PLoS One 2021; 16:e0257254. [PMID: 34582479 PMCID: PMC8478202 DOI: 10.1371/journal.pone.0257254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/26/2021] [Indexed: 11/19/2022] Open
Abstract
Osteogenesis imperfecta (OI) type XIV is a rare recessive bone disorder characterized by variable degree of severity associated to osteopenia. It is caused by mutations in TMEM38B encoding for the trimeric intracellular cation channel TRIC-B, specific for potassium and ubiquitously present in the endoplasmic reticulum (ER) membrane. OI type XIV molecular basis is largely unknown and, due to the rarity of the disease, the availability of patients’ osteoblasts is challenging. Thus, CRISPR/Cas9 was used to knock out (KO) TMEM38B in the human Foetal Osteoblast hFOB 1.19 to obtain an OI type XIV model. CRISPR/Cas9 is a powerful technology to generate in vitro and in vivo models for heritable disorders. Its limited cost and ease of use make this technique widely applicable in most laboratories. Nevertheless, to fully take advantage of this approach, it is important to be aware of its strengths and limitations. Three gRNAs were used and several KO clones lacking the expression of TRIC-B were obtained. Few clones were validated as good models for the disease since they reproduce the altered ER calcium flux, collagen I structure and impaired secretion and osteoblastic markers expression detected in patients’ cells. Impaired proliferation and mineralization in KO clones unveiled the relevance of TRIC-B in osteoblasts functionality.
Collapse
|
9
|
Role of protons in calcium signaling. Biochem J 2021; 478:895-910. [PMID: 33635336 DOI: 10.1042/bcj20200971] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/28/2021] [Accepted: 02/01/2021] [Indexed: 02/03/2023]
Abstract
Thirty-six years after the publication of the important article by Busa and Nuccitelli on the variability of intracellular pH (pHi) and the interdependence of pHi and intracellular Ca2+ concentration ([Ca2+]i), little research has been carried out on pHi and calcium signaling. Moreover, the results appear to be contradictory. Some authors claim that the increase in [Ca2+]i is due to a reduction in pHi, others that it is caused by an increase in pHi. The reasons for these conflicting results have not yet been discussed and clarified in an exhaustive manner. The idea that variations in pHi are insignificant, because cellular buffers quickly stabilize the pHi, may be a limiting and fundamentally wrong concept. In fact, it has been shown that protons can move and react in the cell before they are neutralized. Variations in pHi have a remarkable impact on [Ca2+]i and hence on some of the basic biochemical mechanisms of calcium signaling. This paper focuses on the possible triggering role of protons during their short cellular cycle and it suggests a new hypothesis for an IP3 proton dependent mechanism of action.
Collapse
|
10
|
Gaspers LD, Thomas AP, Hoek JB, Bartlett PJ. Ethanol Disrupts Hormone-Induced Calcium Signaling in Liver. FUNCTION (OXFORD, ENGLAND) 2021; 2:zqab002. [PMID: 33604575 PMCID: PMC7875097 DOI: 10.1093/function/zqab002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/22/2020] [Accepted: 12/30/2020] [Indexed: 01/06/2023]
Abstract
Receptor-coupled phospholipase C (PLC) is an important target for the actions of ethanol. In the ex vivo perfused rat liver, concentrations of ethanol >100 mM were required to induce a rise in cytosolic calcium (Ca2+) suggesting that these responses may only occur after binge ethanol consumption. Conversely, pharmacologically achievable concentrations of ethanol (≤30 mM) decreased the frequency and magnitude of hormone-stimulated cytosolic and nuclear Ca2+ oscillations and the parallel translocation of protein kinase C-β to the membrane. Ethanol also inhibited gap junction communication resulting in the loss of coordinated and spatially organized intercellular Ca2+ waves in hepatic lobules. Increasing the hormone concentration overcame the effects of ethanol on the frequency of Ca2+ oscillations and amplitude of the individual Ca2+ transients; however, the Ca2+ responses in the intact liver remained disorganized at the intercellular level, suggesting that gap junctions were still inhibited. Pretreating hepatocytes with an alcohol dehydrogenase inhibitor suppressed the effects of ethanol on hormone-induced Ca2+ increases, whereas inhibiting aldehyde dehydrogenase potentiated the inhibitory actions of ethanol, suggesting that acetaldehyde is the underlying mediator. Acute ethanol intoxication inhibited the rate of rise and the magnitude of hormone-stimulated production of inositol 1,4,5-trisphosphate (IP3), but had no effect on the size of Ca2+ spikes induced by photolysis of caged IP3. These findings suggest that ethanol inhibits PLC activity, but does not affect IP3 receptor function. We propose that by suppressing hormone-stimulated PLC activity, ethanol interferes with the dynamic modulation of [IP3] that is required to generate large, amplitude Ca2+ oscillations.
Collapse
Affiliation(s)
- Lawrence D Gaspers
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA,Address correspondence to L.D.G. (e-mail: )
| | - Andrew P Thomas
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Jan B Hoek
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Paula J Bartlett
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| |
Collapse
|
11
|
Various Aspects of Calcium Signaling in the Regulation of Apoptosis, Autophagy, Cell Proliferation, and Cancer. Int J Mol Sci 2020; 21:ijms21218323. [PMID: 33171939 PMCID: PMC7664196 DOI: 10.3390/ijms21218323] [Citation(s) in RCA: 182] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/28/2020] [Accepted: 11/03/2020] [Indexed: 12/11/2022] Open
Abstract
Calcium (Ca2+) is a major second messenger in cells and is essential for the fate and survival of all higher organisms. Different Ca2+ channels, pumps, or exchangers regulate variations in the duration and levels of intracellular Ca2+, which may be transient or sustained. These changes are then decoded by an elaborate toolkit of Ca2+-sensors, which translate Ca2+ signal to intracellular operational cell machinery, thereby regulating numerous Ca2+-dependent physiological processes. Alterations to Ca2+ homoeostasis and signaling are often deleterious and are associated with certain pathological states, including cancer. Altered Ca2+ transmission has been implicated in a variety of processes fundamental for the uncontrolled proliferation and invasiveness of tumor cells and other processes important for cancer progression, such as the development of resistance to cancer therapies. Here, we review what is known about Ca2+ signaling and how this fundamental second messenger regulates life and death decisions in the context of cancer, with particular attention directed to cell proliferation, apoptosis, and autophagy. We also explore the intersections of Ca2+ and the therapeutic targeting of cancer cells, summarizing the therapeutic opportunities for Ca2+ signal modulators to improve the effectiveness of current anticancer therapies.
Collapse
|
12
|
Dhyani V, Gare S, Gupta RK, Swain S, Venkatesh K, Giri L. GPCR mediated control of calcium dynamics: A systems perspective. Cell Signal 2020; 74:109717. [PMID: 32711109 PMCID: PMC7375278 DOI: 10.1016/j.cellsig.2020.109717] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 02/09/2023]
Abstract
G-protein coupled receptor (GPCR) mediated calcium (Ca2+)-signaling transduction remains crucial in designing drugs for various complex diseases including neurodegeneration, chronic heart failure as well as respiratory diseases. Although there are several reviews detailing various aspects of Ca2+-signaling such as the role of IP3 receptors and Ca2+-induced-Ca2+-release, none of them provide an integrated view of the mathematical descriptions of GPCR signal transduction and investigations on dose-response curves. This article is the first study in reviewing the network structures underlying GPCR signal transduction that control downstream [Cac2+]-oscillations. The central theme of this paper is to present the biochemical pathways, as well as molecular mechanisms underlying the GPCR-mediated Ca2+-dynamics in order to facilitate a better understanding of how agonist concentration is encoded in Ca2+-signals for Gαq, Gαs, and Gαi/o signaling pathways. Moreover, we present the GPCR targeting drugs that are relevant for treating cardiac, respiratory, and neuro-diseases. The current paper presents the ODE formulation for various models along with the detailed schematics of signaling networks. To provide a systems perspective, we present the network motifs that can provide readers an insight into the complex and intriguing science of agonist-mediated Ca2+-dynamics. One of the features of this review is to pinpoint the interplay between positive and negative feedback loops that are involved in controlling intracellular [Cac2+]-oscillations. Furthermore, we review several examples of dose-response curves obtained from [Cac2+]-spiking for various GPCR pathways. This paper is expected to be useful for pharmacologists and computational biologists for designing clinical applications of GPCR targeting drugs through modulation of Ca2+-dynamics.
Collapse
Affiliation(s)
- Vaibhav Dhyani
- Department of Chemical Engineering, Indian Institute of Technology Hyderabad, Sangareddy, Telangana, India
| | - Suman Gare
- Department of Chemical Engineering, Indian Institute of Technology Hyderabad, Sangareddy, Telangana, India
| | - Rishikesh Kumar Gupta
- Department of Chemical Engineering, Indian Institute of Technology Hyderabad, Sangareddy, Telangana, India
| | - Sarpras Swain
- Department of Chemical Engineering, Indian Institute of Technology Hyderabad, Sangareddy, Telangana, India
| | - K.V. Venkatesh
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, India
| | - Lopamudra Giri
- Department of Chemical Engineering, Indian Institute of Technology Hyderabad, Sangareddy, Telangana, India.
| |
Collapse
|
13
|
Dual mechanisms of Ca2+ oscillations in hepatocytes. J Theor Biol 2020; 503:110390. [DOI: 10.1016/j.jtbi.2020.110390] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/19/2020] [Accepted: 06/24/2020] [Indexed: 11/30/2022]
|
14
|
Zhou A, Liu X, Zhang S, Huo B. Effects of store-operated and receptor-operated calcium channels on synchronization of calcium oscillations in astrocytes. Biosystems 2020; 198:104233. [PMID: 32858094 DOI: 10.1016/j.biosystems.2020.104233] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/04/2020] [Accepted: 08/20/2020] [Indexed: 10/23/2022]
Abstract
Intercellular calcium signaling allows cells to communicate with each other and to interact with adjacent cells. Gap junction is the most common and important way for cellular communication. Recently, mathematical models have been widely used to gain a precise and quantitative understanding of the dynamics of intracellular calcium ions (Ca2+). In this paper, we establish a mathematical model considering the gap junction permeable to Ca2+ and to IP3 for describing the calcium oscillations in coupled astrocytes. Store-operated calcium entry (SOCE) is viewed as the main process which controls the non-excitable cells, hence, we focus on the effect of store-operated calcium channel (SOCC) and receptor-operated calcium channel (ROCC) on the intercellular synchronization, respectively. By employing bifurcation analysis on this model, the dynamic behaviors of the coupled system with different physiological state cells is obtained with changes in the maximum capacity of the SOCC and the ROCC. The synchronization boundaries for different conditions are gained in the two parameters space of the channel parameters and the coupling strength. The results suggest that the variation of the maximum flow for different calcium channels determines the stable oscillations of the coupled system, as well as for the frequency and amplitude of oscillations. The SOCC has an expected effect on the change of the oscillatory interval while the ROCC demonstrated the influence on the amplitude modulation. Furthermore, the coupling strength and channel parameters could induce 1:1 locking of intercellular Ca2+ oscillations and the synchronization region like Arnol'd tongue is found.
Collapse
Affiliation(s)
- Anqi Zhou
- Department of Mechanics, Tianjin University, Tianjin, 300354, PR China
| | - Xijun Liu
- Department of Mechanics, Tianjin University, Tianjin, 300354, PR China
| | - Suxia Zhang
- Department of Mechanics, Tianjin University, Tianjin, 300354, PR China.
| | - Bing Huo
- College of Mechanical Engineering, Tianjin University of Science & Technology, Tianjin, 300222, PR China
| |
Collapse
|
15
|
Bartlett PJ, Cloete I, Sneyd J, Thomas AP. IP 3-Dependent Ca 2+ Oscillations Switch into a Dual Oscillator Mechanism in the Presence of PLC-Linked Hormones. iScience 2020; 23:101062. [PMID: 32353764 PMCID: PMC7191650 DOI: 10.1016/j.isci.2020.101062] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 12/11/2019] [Accepted: 04/09/2020] [Indexed: 11/28/2022] Open
Abstract
Ca2+ oscillations that depend on inositol-1,4,5-trisphosphate (IP3) have been ascribed to biphasic Ca2+ regulation of the IP3 receptor (IP3R) or feedback mechanisms controlling IP3 levels in different cell types. IP3 uncaging in hepatocytes elicits Ca2+ transients that are often localized at the subcellular level and increase in magnitude with stimulus strength. However, this does not reproduce the broad baseline-separated global Ca2+ oscillations elicited by vasopressin. Addition of hormone to cells activated by IP3 uncaging initiates a qualitative transition from high-frequency spatially disorganized Ca2+ transients, to low-frequency, oscillatory Ca2+ waves that propagate throughout the cell. A mathematical model with dual coupled oscillators that integrates Ca2+-induced Ca2+ release at the IP3R and mutual feedback mechanisms of cross-coupling between Ca2+ and IP3 reproduces this behavior. Thus, multiple Ca2+ oscillation modes can coexist in the same cell, and hormonal stimulation can switch from the simpler to the more complex to yield robust signaling.
Collapse
Affiliation(s)
- Paula J Bartlett
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Ielyaas Cloete
- Department of Mathematics, The University of Auckland, Auckland, New Zealand
| | - James Sneyd
- Department of Mathematics, The University of Auckland, Auckland, New Zealand
| | - Andrew P Thomas
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School Rutgers, The State University of New Jersey, Newark, NJ 07103, USA.
| |
Collapse
|
16
|
Powell J, Falcke M, Skupin A, Bellamy TC, Kypraios T, Thul R. A Statistical View on Calcium Oscillations. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:799-826. [PMID: 31646535 DOI: 10.1007/978-3-030-12457-1_32] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Transient rises and falls of the intracellular calcium concentration have been observed in numerous cell types and under a plethora of conditions. There is now a growing body of evidence that these whole-cell calcium oscillations are stochastic, which poses a significant challenge for modelling. In this review, we take a closer look at recently developed statistical approaches to calcium oscillations. These models describe the timing of whole-cell calcium spikes, yet their parametrisations reflect subcellular processes. We show how non-stationary calcium spike sequences, which e.g. occur during slow depletion of intracellular calcium stores or in the presence of time-dependent stimulation, can be analysed with the help of so-called intensity functions. By utilising Bayesian concepts, we demonstrate how values of key parameters of the statistical model can be inferred from single cell calcium spike sequences and illustrate what information whole-cell statistical models can provide about the subcellular mechanistic processes that drive calcium oscillations. In particular, we find that the interspike interval distribution of HEK293 cells under constant stimulation is captured by a Gamma distribution.
Collapse
Affiliation(s)
- Jake Powell
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, UK
| | - Martin Falcke
- Max Delbrück Centre for Molecular Medicine, Berlin, Germany.,Department of Physics, Humboldt University, Berlin, Germany
| | - Alexander Skupin
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg.,National Biomedical Computation Resource, University California San Diego, La Jolla, CA, USA
| | - Tomas C Bellamy
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Theodore Kypraios
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, UK
| | - Rüdiger Thul
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, UK.
| |
Collapse
|
17
|
Porcine deltacoronavirus (PDCoV) modulates calcium influx to favor viral replication. Virology 2019; 539:38-48. [PMID: 31670218 PMCID: PMC7112098 DOI: 10.1016/j.virol.2019.10.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/12/2022]
Abstract
Ionic calcium (Ca2+) is a versatile intracellular second messenger that plays important roles in cellular physiological and pathological processes. Porcine deltacoronavirus (PDCoV) is an emerging enteropathogenic coronavirus that causes serious vomiting and diarrhea in suckling piglets. In this study, the role of Ca2+ to PDCoV infection was investigated. PDCoV infection was found to upregulate intracellular Ca2+ concentrations of IPI-2I cells. Chelating extracellular Ca2+ by EGTA inhibited PDCoV replication, and this inhibitory effect was overcome by replenishment with CaCl2. Treatment with Ca2+ channel blockers, particularly the L-type Ca2+ channel blocker diltiazem hydrochloride, inhibited PDCoV infection significantly. Mechanistically, diltiazem hydrochloride reduces PDCoV infection by inhibiting the replication step of the viral replication cycle. Additionally, knockdown of CACNA1S, the L-type Ca2+ voltage-gated channel subunit, inhibited PDCoV replication. The combined results demonstrate that PDCoV modulates calcium influx to favor its replication.
Collapse
|
18
|
Pages N, Vera-Sigüenza E, Rugis J, Kirk V, Yule DI, Sneyd J. A Model of [Formula: see text] Dynamics in an Accurate Reconstruction of Parotid Acinar Cells. Bull Math Biol 2019; 81:1394-1426. [PMID: 30644065 PMCID: PMC6449190 DOI: 10.1007/s11538-018-00563-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 12/21/2018] [Indexed: 01/23/2023]
Abstract
We have constructed a spatiotemporal model of [Formula: see text] dynamics in parotid acinar cells, based on new data about the distribution of inositol trisphophate receptors (IPR). The model is solved numerically on a mesh reconstructed from images of a cluster of parotid acinar cells. In contrast to our earlier model (Sneyd et al. in J Theor Biol 419:383-393. https://doi.org/10.1016/j.jtbi.2016.04.030 , 2017b), which cannot generate realistic [Formula: see text] oscillations with the new data on IPR distribution, our new model reproduces the [Formula: see text] dynamics observed in parotid acinar cells. This model is then coupled with a fluid secretion model described in detail in a companion paper: A mathematical model of fluid transport in an accurate reconstruction of a parotid acinar cell (Vera-Sigüenza et al. in Bull Math Biol. https://doi.org/10.1007/s11538-018-0534-z , 2018b). Based on the new measurements of IPR distribution, we show that Class I models (where [Formula: see text] oscillations can occur at constant [[Formula: see text]]) can produce [Formula: see text] oscillations in parotid acinar cells, whereas Class II models (where [[Formula: see text]] needs to oscillate in order to produce [Formula: see text] oscillations) are unlikely to do so. In addition, we demonstrate that coupling fluid flow secretion with the [Formula: see text] signalling model changes the dynamics of the [Formula: see text] oscillations significantly, which indicates that [Formula: see text] dynamics and fluid flow cannot be accurately modelled independently. Further, we determine that an active propagation mechanism based on calcium-induced calcium release channels is needed to propagate the [Formula: see text] wave from the apical region to the basal region of the acinar cell.
Collapse
Affiliation(s)
- Nathan Pages
- Department of Mathematics, The University of Auckland, 38 Princes Street, Auckland 1010, New Zealand
| | - Elías Vera-Sigüenza
- Department of Mathematics, The University of Auckland, 38 Princes Street, Auckland 1010, New Zealand
| | - John Rugis
- Department of Mathematics, The University of Auckland, 38 Princes Street, Auckland 1010, New Zealand
| | - Vivien Kirk
- Department of Mathematics, The University of Auckland, 38 Princes Street, Auckland 1010, New Zealand
| | - David I. Yule
- University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, Box 711, Rochester NY, United States of America
| | - James Sneyd
- Department of Mathematics, The University of Auckland, 38 Princes Street, Auckland 1010, New Zealand
| |
Collapse
|
19
|
Gaspers LD, Pierobon N, Thomas AP. Intercellular calcium waves integrate hormonal control of glucose output in the intact liver. J Physiol 2019; 597:2867-2885. [PMID: 30968953 PMCID: PMC6647271 DOI: 10.1113/jp277650] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/08/2019] [Indexed: 11/21/2022] Open
Abstract
Key points Sympathetic outflow and circulating glucogenic hormones both regulate liver function by increasing cytosolic calcium, although how these calcium signals are integrated at the tissue level is currently unknown. We show that stimulation of hepatic nerve fibres or perfusing the liver with physiological concentrations of vasopressin only will evoke localized cytosolic calcium oscillations and modest increases in hepatic glucose production. The combination of these stimuli acted synergistically to convert localized and asynchronous calcium responses into co‐ordinated intercellular calcium waves that spread throughout the liver lobule and elicited a synergistic increase in hepatic glucose production. The results obtained in the present study demonstrate that subthreshold levels of one hormone can create an excitable medium across the liver lobule, which allows global propagation of calcium signals in response to local sympathetic innervation and integration of metabolic regulation by multiple hormones. This enables the liver lobules to respond as functional units to produce full‐strength metabolic output at physiological levels of hormone.
Abstract Glucogenic hormones, including catecholamines and vasopressin, induce frequency‐modulated cytosolic Ca2+ oscillations in hepatocytes, and these propagate as intercellular Ca2+ waves via gap junctions in the intact liver. We investigated the role of co‐ordinated Ca2+ waves as a mechanism for integrating multiple endocrine and neuroendocrine inputs to control hepatic glucose production in perfused rat liver. Sympathetic nerve stimulation elicited localized Ca2+ increases that were restricted to hepatocytes in the periportal zone. During perfusion with subthreshold vasopressin, sympathetic stimulation converted asynchronous Ca2+ signals in a limited number of hepatocytes into co‐ordinated intercellular Ca2+ waves that propagated across entire lobules. A similar synergism was observed between physiological concentrations of glucagon and vasopressin, where glucagon also facilitated the recruitment of hepatocytes into a Ca2+ wave. Hepatic glucose production was significantly higher with intralobular Ca2+ waves. We propose that inositol 1,4,5‐trisphosphate (IP3)‐dependent Ca2+ signalling gives rise to an excitable medium across the functional syncytium of the hepatic lobule, co‐ordinating and amplifying the metabolic responses to multiple hormonal inputs. Sympathetic outflow and circulating glucogenic hormones both regulate liver function by increasing cytosolic calcium, although how these calcium signals are integrated at the tissue level is currently unknown. We show that stimulation of hepatic nerve fibres or perfusing the liver with physiological concentrations of vasopressin only will evoke localized cytosolic calcium oscillations and modest increases in hepatic glucose production. The combination of these stimuli acted synergistically to convert localized and asynchronous calcium responses into co‐ordinated intercellular calcium waves that spread throughout the liver lobule and elicited a synergistic increase in hepatic glucose production. The results obtained in the present study demonstrate that subthreshold levels of one hormone can create an excitable medium across the liver lobule, which allows global propagation of calcium signals in response to local sympathetic innervation and integration of metabolic regulation by multiple hormones. This enables the liver lobules to respond as functional units to produce full‐strength metabolic output at physiological levels of hormone.
Collapse
Affiliation(s)
- Lawrence D Gaspers
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Nicola Pierobon
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Andrew P Thomas
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School Rutgers, The State University of New Jersey, Newark, NJ, USA
| |
Collapse
|
20
|
Wacquier B, Voorsluijs V, Combettes L, Dupont G. Coding and decoding of oscillatory Ca 2+ signals. Semin Cell Dev Biol 2019; 94:11-19. [PMID: 30659886 DOI: 10.1016/j.semcdb.2019.01.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/11/2019] [Accepted: 01/14/2019] [Indexed: 01/08/2023]
Abstract
About 30 years after their first observation, Ca2+ oscillations are now recognised as a universal mechanism of signal transduction. These oscillations are driven by periodic cycles of release and uptake of Ca2+ between the cytoplasm and the endoplasmic reticulum. Their frequency often increases with the level of stimulation, which can be decoded by some molecules. However, it is becoming increasingly evident that the widespread core oscillatory mechanism is modulated in many ways, depending on the cell type and on the physiological conditions. Interplay with inositol 1,4,5-trisphosphate metabolism and with other Ca2+ stores as the extracellular medium or mitochondria can much affect the properties of these oscillations. In many cases, these finely tuned characteristics of Ca2+ oscillations impact the physiological response that is triggered by the signal. Moreover, oscillations are intrinsically irregular. This randomness can also be exploited by the cell. In this review, we discuss evidences of these additional manifestations of the versatility of Ca2+ signalling.
Collapse
Affiliation(s)
- Benjamin Wacquier
- Unit of Theoretical Chronobiology, Faculté des Sciences, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Valérie Voorsluijs
- Nonlinear Physical Chemistry Unit & Center for Nonlinear Phenomena and Complex Systems (CENOLI), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - Geneviève Dupont
- Unit of Theoretical Chronobiology, Faculté des Sciences, Université Libre de Bruxelles (ULB), Brussels, Belgium.
| |
Collapse
|
21
|
Shears SB, Wang H. Inositol phosphate kinases: Expanding the biological significance of the universal core of the protein kinase fold. Adv Biol Regul 2019; 71:118-127. [PMID: 30392847 PMCID: PMC9364425 DOI: 10.1016/j.jbior.2018.10.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 10/23/2018] [Accepted: 10/26/2018] [Indexed: 05/06/2023]
Abstract
The protein kinase family is characterized by substantial conservation of architectural elements that are required for both ATP binding and phosphotransferase activity. Many of these structural features have also been identified in homologous enzymes that phosphorylate a variety of alternative, non-protein substrates. A comparative structural analysis of these different kinase sub-classes is a portal to a greater understanding of reaction mechanisms, enzyme regulation, inhibitor-development strategies, and superfamily-level evolutionary relationships. To serve such advances, we review structural elements of the protein kinase fold that are conserved in the subfamily of inositol phosphate kinases (InsPKs) that share a PxxxDxKxG catalytic signature: inositol 1,4,5-trisphosphate kinase (IP3K), inositol hexakisphosphate kinase (IP6K), and inositol polyphosphate multikinase (IPMK). We describe conservation of the fundamental two-lobe kinase architecture: an N-lobe constructed upon an anti-parallel β-strand scaffold, which is coupled to a largely helical C-lobe by a single, adenine-binding hinge. This equivalency also includes a G-loop that embraces the β/γ-phosphates of ATP, a transition-state stabilizing residue (Lys/His), and a Mg-positioning aspartate residue within a catalytic triad. Furthermore, we expand this list of conserved structural features to include some not previously identified in InsPKs: a 'gatekeeper' residue in the N-lobe, and an 'αF'-like helix in the C-lobe that anchors two structurally-stabilizing, hydrophobic spines, formed from non-consecutive residues that span the two lobes. We describe how this wide-ranging structural homology can be exploited to develop lead inhibitors of IP6K and IPMK, by using strategies similar to those that have generated ATP-competing inhibitors of protein-kinases. We provide several examples to illustrate how such an approach could benefit human health.
Collapse
Affiliation(s)
- Stephen B Shears
- Inositol Signaling Group, Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA.
| | - Huanchen Wang
- Inositol Signaling Group, Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA.
| |
Collapse
|
22
|
Shimobayashi E, Kapfhammer JP. Calcium Signaling, PKC Gamma, IP3R1 and CAR8 Link Spinocerebellar Ataxias and Purkinje Cell Dendritic Development. Curr Neuropharmacol 2018; 16:151-159. [PMID: 28554312 PMCID: PMC5883377 DOI: 10.2174/1570159x15666170529104000] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 05/16/2017] [Accepted: 05/25/2017] [Indexed: 01/05/2023] Open
Abstract
Background Spinocerebellar ataxias (SCAs) are a group of cerebellar diseases characterized by progressive ataxia and cerebellar atrophy. Several forms of SCAs are caused by missense mutations or deletions in genes related to calcium signaling in Purkinje cells. Among them, spinocerebellar ataxia type 14 (SCA14) is caused by missense mutations in PRKCG gene which encodes protein kinase C gamma (PKCγ). It is remarkable that in several cases in which SCA is caused by point mutations in an individual gene, the affected genes are involved in the PKCγ signaling pathway and calcium signaling which is not only crucial for proper Purkinje cell function but is also involved in the control of Purkinje cell dendritic development. In this review, we will focus on the PKCγ signaling related genes and calcium signaling related genes then discuss their role for both Purkinje cell dendritic development and cerebellar ataxia. Methods Research related to SCAs and Purkinje cell dendritic development is reviewed. Results PKCγ dysregulation causes abnormal Purkinje cell dendritic development and SCA14. Carbonic anhydrase related protein 8 (Car8) encoding CAR8 and Itpr1 encoding IP3R1were identified as upregulated genes in one of SCA14 mouse model. IP3R1, CAR8 and PKCγ proteins are strongly and specifically expressed in Purkinje cells. The common function among them is that they are involved in the regulation of calcium homeostasis in Purkinje cells and their dysfunction causes ataxia in mouse and human. Furthermore, disruption of intracellular calcium homeostasis caused by mutations in some calcium channels in Purkinje cells links to abnormal Purkinje cell dendritic development and the pathogenesis of several SCAs. Conclusion Once PKCγ signaling related genes and calcium signaling related genes are disturbed, the normal dendritic development of Purkinje cells is impaired as well as the integration of signals from other neurons, resulting in abnormal development, cerebellar dysfunction and eventually Purkinje cell loss.
Collapse
Affiliation(s)
- Etsuko Shimobayashi
- Anatomical Institute, Department of Biomedicine Basel, University of Basel, Pestalozzistrasse 20, CH-4056 Basel, Switzerland
| | - Josef P Kapfhammer
- Anatomical Institute, Department of Biomedicine Basel, University of Basel, Pestalozzistrasse 20, CH-4056 Basel, Switzerland
| |
Collapse
|
23
|
Role of KCa3.1 Channels in Modulating Ca 2+ Oscillations during Glioblastoma Cell Migration and Invasion. Int J Mol Sci 2018; 19:ijms19102970. [PMID: 30274242 PMCID: PMC6213908 DOI: 10.3390/ijms19102970] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/21/2018] [Accepted: 09/25/2018] [Indexed: 01/29/2023] Open
Abstract
Cell migration and invasion in glioblastoma (GBM), the most lethal form of primary brain tumors, are critically dependent on Ca2+ signaling. Increases of [Ca2+]i in GBM cells often result from Ca2+ release from the endoplasmic reticulum (ER), promoted by a variety of agents present in the tumor microenvironment and able to activate the phospholipase C/inositol 1,4,5-trisphosphate PLC/IP3 pathway. The Ca2+ signaling is further strengthened by the Ca2+ influx from the extracellular space through Ca2+ release-activated Ca2+ (CRAC) currents sustained by Orai/STIM channels, meant to replenish the partially depleted ER. Notably, the elevated cytosolic [Ca2+]i activates the intermediate conductance Ca2+-activated K (KCa3.1) channels highly expressed in the plasma membrane of GBM cells, and the resulting K+ efflux hyperpolarizes the cell membrane. This translates to an enhancement of Ca2+ entry through Orai/STIM channels as a result of the increased electromotive (driving) force on Ca2+ influx, ending with the establishment of a recurrent cycle reinforcing the Ca2+ signal. Ca2+ signaling in migrating GBM cells often emerges in the form of intracellular Ca2+ oscillations, instrumental to promote key processes in the migratory cycle. This has suggested that KCa3.1 channels may promote GBM cell migration by inducing or modulating the shape of Ca2+ oscillations. In accordance, we recently built a theoretical model of Ca2+ oscillations incorporating the KCa3.1 channel-dependent dynamics of the membrane potential, and found that the KCa3.1 channel activity could significantly affect the IP3 driven Ca2+ oscillations. Here we review our new theoretical model of Ca2+ oscillations in GBM, upgraded in the light of better knowledge of the KCa3.1 channel kinetics and Ca2+ sensitivity, the dynamics of the Orai/STIM channel modulation, the migration and invasion mechanisms of GBM cells, and their regulation by Ca2+ signals.
Collapse
|
24
|
Watson U, Jain R, Asthana S, Saini DK. Spatiotemporal Modulation of ERK Activation by GPCRs. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 338:111-140. [DOI: 10.1016/bs.ircmb.2018.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
|
25
|
Grundmann M, Kostenis E. Temporal Bias: Time-Encoded Dynamic GPCR Signaling. Trends Pharmacol Sci 2017; 38:1110-1124. [PMID: 29074251 DOI: 10.1016/j.tips.2017.09.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 09/24/2017] [Accepted: 09/25/2017] [Indexed: 12/21/2022]
Abstract
Evidence suggests that cells can time-encode signals for secure transport and perception of information, and it appears that this dynamic signaling is a common principle of nature to code information in time. G-protein-coupled receptor (GPCR) signaling networks are no exception as their composition and signal transduction appear temporally flexible. In this review, we discuss the potential mechanisms by which GPCRs code biological information in time to create 'temporal bias.' We highlight dynamic signaling patterns from the second messenger to the receptor-ligand level and shed light on the dynamics of G-protein cycles, the kinetics of ligand-receptor interaction, and the occurrence of distinct signaling waves within the cell. A dynamic feature such as temporal bias adds to the complexity of GPCR signaling bias and gives rise to the question whether this trait could be exploited to gain control over time-encoded cell physiology.
Collapse
Affiliation(s)
- Manuel Grundmann
- Molecular-, Cellular- and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany; Kidney Disease Research, Bayer Pharma AG, Aprather Weg 18a, 42113 Wuppertal, Germany
| | - Evi Kostenis
- Molecular-, Cellular- and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany.
| |
Collapse
|
26
|
Lees JA, Messa M, Sun EW, Wheeler H, Torta F, Wenk MR, De Camilli P, Reinisch KM. Lipid transport by TMEM24 at ER-plasma membrane contacts regulates pulsatile insulin secretion. Science 2017; 355:355/6326/eaah6171. [PMID: 28209843 DOI: 10.1126/science.aah6171] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 01/03/2017] [Indexed: 01/10/2023]
Abstract
Insulin is released by β cells in pulses regulated by calcium and phosphoinositide signaling. Here, we describe how transmembrane protein 24 (TMEM24) helps coordinate these signaling events. We showed that TMEM24 is an endoplasmic reticulum (ER)-anchored membrane protein whose reversible localization to ER-plasma membrane (PM) contacts is governed by phosphorylation and dephosphorylation in response to oscillations in cytosolic calcium. A lipid-binding module in TMEM24 transports the phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] precursor phosphatidylinositol between bilayers, allowing replenishment of PI(4,5)P2 hydrolyzed during signaling. In the absence of TMEM24, calcium oscillations are abolished, leading to a defect in triggered insulin release. Our findings implicate direct lipid transport between the ER and the PM in the control of insulin secretion, a process impaired in patients with type II diabetes.
Collapse
Affiliation(s)
- Joshua A Lees
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Mirko Messa
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA.,Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA.,Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA.,Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Elizabeth Wen Sun
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA.,Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA.,Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA.,Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Heather Wheeler
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA.,Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA.,Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA.,Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Federico Torta
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore
| | - Markus R Wenk
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore
| | - Pietro De Camilli
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA. .,Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA.,Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA.,Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT 06510, USA.,Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Karin M Reinisch
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
27
|
Bartlett PJ, Antony AN, Agarwal A, Hilly M, Prince VL, Combettes L, Hoek JB, Gaspers LD. Chronic alcohol feeding potentiates hormone-induced calcium signalling in hepatocytes. J Physiol 2017; 595:3143-3164. [PMID: 28220501 DOI: 10.1113/jp273891] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 01/26/2017] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS Chronic alcohol consumption causes a spectrum of liver diseases, but the pathogenic mechanisms driving the onset and progression of disease are not clearly defined. We show that chronic alcohol feeding sensitizes rat hepatocytes to Ca2+ -mobilizing hormones resulting in a leftward shift in the concentration-response relationship and the transition from oscillatory to more sustained and prolonged Ca2+ increases. Our data demonstrate that alcohol-dependent adaptation in the Ca2+ signalling pathway occurs at the level of hormone-induced inositol 1,4,5 trisphosphate (IP3 ) production and does not involve changes in the sensitivity of the IP3 receptor or size of internal Ca2+ stores. We suggest that prolonged and aberrant hormone-evoked Ca2+ increases may stimulate the production of mitochondrial reactive oxygen species and contribute to alcohol-induced hepatocyte injury. ABSTRACT: 'Adaptive' responses of the liver to chronic alcohol consumption may underlie the development of cell and tissue injury. Alcohol administration can perturb multiple signalling pathways including phosphoinositide-dependent cytosolic calcium ([Ca2+ ]i ) increases, which can adversely affect mitochondrial Ca2+ levels, reactive oxygen species production and energy metabolism. Our data indicate that chronic alcohol feeding induces a leftward shift in the dose-response for Ca2+ -mobilizing hormones resulting in more sustained and prolonged [Ca2+ ]i increases in both cultured hepatocytes and hepatocytes within the intact perfused liver. Ca2+ increases were initiated at lower hormone concentrations, and intercellular calcium wave propagation rates were faster in alcoholics compared to controls. Acute alcohol treatment (25 mm) completely inhibited hormone-induced calcium increases in control livers, but not after chronic alcohol-feeding, suggesting desensitization to the inhibitory actions of ethanol. Hormone-induced inositol 1,4,5 trisphosphate (IP3 ) accumulation and phospholipase C (PLC) activity were significantly potentiated in hepatocytes from alcohol-fed rats compared to controls. Removal of extracellular calcium, or chelation of intracellular calcium did not normalize the differences in hormone-stimulated PLC activity, indicating calcium-dependent PLCs are not upregulated by alcohol. We propose that the liver 'adapts' to chronic alcohol exposure by increasing hormone-dependent IP3 formation, leading to aberrant calcium increases, which may contribute to hepatocyte injury.
Collapse
Affiliation(s)
- Paula J Bartlett
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - Anil Noronha Antony
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Amit Agarwal
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - Mauricette Hilly
- INSERM UMR-S 757, Université de Paris-Sud, bât 443, 91405, Orsay, France
| | - Victoria L Prince
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - Laurent Combettes
- INSERM UMR-S 757, Université de Paris-Sud, bât 443, 91405, Orsay, France
| | - Jan B Hoek
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Lawrence D Gaspers
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| |
Collapse
|
28
|
Han JM, Tanimura A, Kirk V, Sneyd J. A mathematical model of calcium dynamics in HSY cells. PLoS Comput Biol 2017; 13:e1005275. [PMID: 28199326 PMCID: PMC5310762 DOI: 10.1371/journal.pcbi.1005275] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 11/30/2016] [Indexed: 12/03/2022] Open
Abstract
Saliva is an essential part of activities such as speaking, masticating and swallowing. Enzymes in salivary fluid protect teeth and gums from infectious diseases, and also initiate the digestion process. Intracellular calcium (Ca2+) plays a critical role in saliva secretion and regulation. Experimental measurements of Ca2+ and inositol trisphosphate (IP3) concentrations in HSY cells, a human salivary duct cell line, show that when the cells are stimulated with adenosine triphosphate (ATP) or carbachol (CCh), they exhibit coupled oscillations with Ca2+ spike peaks preceding IP3 spike peaks. Based on these data, we construct a mathematical model of coupled Ca2+ and IP3 oscillations in HSY cells and perform model simulations of three different experimental settings to forecast Ca2+ responses. The model predicts that when Ca2+ influx from the extracellular space is removed, oscillations gradually slow down until they stop. The model simulation of applying a pulse of IP3 predicts that photolysis of caged IP3 causes a transient increase in the frequency of the Ca2+ oscillations. Lastly, when Ca2+-dependent activation of PLC is inhibited, we see an increase in the oscillation frequency and a decrease in the amplitude. These model predictions are confirmed by experimental data. We conclude that, although concentrations of Ca2+ and IP3 oscillate, Ca2+ oscillations in HSY cells are the result of modulation of the IP3 receptor by intracellular Ca2+, and that the period is modulated by the accompanying IP3 oscillations.
Collapse
Affiliation(s)
- Jung Min Han
- Department of Mathematics, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Akihiko Tanimura
- Department of Pharmacology, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido 061-0293, Japan
| | - Vivien Kirk
- Department of Mathematics, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - James Sneyd
- Department of Mathematics, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
29
|
Abstract
Oscillations in the concentration of free cytosolic Ca2+ are an important and ubiquitous control mechanism in many cell types. It is thus correspondingly important to understand the mechanisms that underlie the control of these oscillations and how their period is determined. We show that Class I Ca2+ oscillations (i.e., oscillations that can occur at a constant concentration of inositol trisphosphate) have a common dynamical structure, irrespective of the oscillation period. This commonality allows the construction of a simple canonical model that incorporates this underlying dynamical behavior. Predictions from the model are tested, and confirmed, in three different cell types, with oscillation periods ranging over an order of magnitude. The model also predicts that Ca2+ oscillation period can be controlled by modulation of the rate of activation by Ca2+ of the inositol trisphosphate receptor. Preliminary experimental evidence consistent with this hypothesis is presented. Our canonical model has a structure similar to, but not identical to, the classic FitzHugh-Nagumo model. The characterization of variables by speed of evolution, as either fast or slow variables, changes over the course of a typical oscillation, leading to a model without globally defined fast and slow variables.
Collapse
|
30
|
Miyamoto A, Mikoshiba K. Probes for manipulating and monitoring IP 3. Cell Calcium 2016; 64:57-64. [PMID: 27887748 DOI: 10.1016/j.ceca.2016.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 11/14/2016] [Indexed: 12/31/2022]
Abstract
Inositol 1,4,5-trisphosphate (IP3) is an important second messenger produced via G-protein-coupled receptor- or receptor tyrosine kinase-mediated pathways. IP3 levels induce Ca2+ release from the endoplasmic reticulum (ER) via IP3 receptor (IP3R) located in the ER membrane. The resultant spatiotemporal pattern of Ca2+ signals regulates diverse cellular functions, including fertilization, gene expression, synaptic plasticity, and cell death. Therefore, monitoring and manipulating IP3 levels is important to elucidate not only the functions of IP3-mediated pathways but also the encoding mechanism of IP3R as a converter of intracellular signals from IP3 to Ca2+.
Collapse
Affiliation(s)
- Akitoshi Miyamoto
- Laboratory for Developmental Neurobiology, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Katsuhiko Mikoshiba
- Laboratory for Developmental Neurobiology, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan.
| |
Collapse
|
31
|
Critical role of ATP-induced ATP release for Ca2+ signaling in nonsensory cell networks of the developing cochlea. Proc Natl Acad Sci U S A 2016; 113:E7194-E7201. [PMID: 27807138 DOI: 10.1073/pnas.1616061113] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Spatially and temporally coordinated variations of the cytosolic free calcium concentration ([Ca2+]c) play a crucial role in a variety of tissues. In the developing sensory epithelium of the mammalian cochlea, elevation of extracellular adenosine trisphosphate concentration ([ATP]e) triggers [Ca2+]c oscillations and propagation of intercellular inositol 1,4,5-trisphosphate (IP3)-dependent Ca2+ waves. What remains uncertain is the relative contribution of gap junction channels and connexin hemichannels to these fundamental mechanisms, defects in which impair hearing acquisition. Another related open question is whether [Ca2+]c oscillations require oscillations of the cytosolic IP3 concentration ([IP3]c) in this system. To address these issues, we performed Ca2+ imaging experiments in the lesser epithelial ridge of the mouse cochlea around postnatal day 5 and constructed a computational model in quantitative adherence to experimental data. Our results indicate that [Ca2+]c oscillations are governed by Hopf-type bifurcations within the experimental range of [ATP]e and do not require [IP3]c oscillations. The model replicates accurately the spatial extent and propagation speed of intercellular Ca2+ waves and predicts that ATP-induced ATP release is the primary mechanism underlying intercellular propagation of Ca2+ signals. The model also uncovers a discontinuous transition from propagating regimes (intercellular Ca2+ wave speed > 11 μm⋅s-1) to propagation failure (speed = 0), which occurs upon lowering the maximal ATP release rate below a minimal threshold value. The approach presented here overcomes major limitations due to lack of specific connexin channel inhibitors and can be extended to other coupled cellular systems.
Collapse
|
32
|
Rückl M, Rüdiger S. Calcium waves in a grid of clustered channels with synchronous IP 3 binding and unbinding. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2016; 39:108. [PMID: 27848113 DOI: 10.1140/epje/i2016-16108-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 10/26/2016] [Indexed: 06/06/2023]
Abstract
Calcium signals in cells occur at multiple spatial scales and variable temporal duration. However, a physical explanation for transitions between long-lasting global oscillations and localized short-term elevations (puffs) of cytoplasmic Ca2+ is still lacking. Here we introduce a phenomenological, coarse-grained model for the calcium variable, which is represented by ordinary differential equations. Due to its small number of parameters, and its simplicity, this model allows us to numerically study the interplay of multi-scale calcium concentrations with stochastic ion channel gating dynamics even in larger systems. We apply this model to a single cluster of inositol trisphosphate (IP 3) receptor channels and find further evidence for the results presented in earlier work: a single cluster may be capable of producing different calcium release types, where long-lasting events are accompanied by unbinding of IP 3 from the receptor (Rückl et al., PLoS Comput. Biol. 11, e1003965 (2015)). Finally, we show the practicability of the model in a grid of 64 clusters which is computationally intractable with previous high-resolution models. Here long-lasting events can lead to synchronized oscillations and waves, while short events stay localized. The frequency of calcium releases as well as their coherence can thereby be regulated by the amplitude of IP 3 stimulation. Finally the model allows for a new explanation of oscillating [IP 3], which is not based on metabolic production and degradation of IP 3.
Collapse
Affiliation(s)
- M Rückl
- Institut für Physik, Humboldt-Universität zu Berlin, Berlin, Germany.
| | - S Rüdiger
- Institut für Physik, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
33
|
Berridge MJ. The Inositol Trisphosphate/Calcium Signaling Pathway in Health and Disease. Physiol Rev 2016; 96:1261-96. [DOI: 10.1152/physrev.00006.2016] [Citation(s) in RCA: 377] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Many cellular functions are regulated by calcium (Ca2+) signals that are generated by different signaling pathways. One of these is the inositol 1,4,5-trisphosphate/calcium (InsP3/Ca2+) signaling pathway that operates through either primary or modulatory mechanisms. In its primary role, it generates the Ca2+ that acts directly to control processes such as metabolism, secretion, fertilization, proliferation, and smooth muscle contraction. Its modulatory role occurs in excitable cells where it modulates the primary Ca2+ signal generated by the entry of Ca2+ through voltage-operated channels that releases Ca2+ from ryanodine receptors (RYRs) on the internal stores. In carrying out this modulatory role, the InsP3/Ca2+ signaling pathway induces subtle changes in the generation and function of the voltage-dependent primary Ca2+ signal. Changes in the nature of both the primary and modulatory roles of InsP3/Ca2+ signaling are a contributory factor responsible for the onset of a large number human diseases.
Collapse
Affiliation(s)
- Michael J. Berridge
- Laboratory of Molecular Signalling, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, United Kingdom
| |
Collapse
|
34
|
Raffaello A, Mammucari C, Gherardi G, Rizzuto R. Calcium at the Center of Cell Signaling: Interplay between Endoplasmic Reticulum, Mitochondria, and Lysosomes. Trends Biochem Sci 2016; 41:1035-1049. [PMID: 27692849 DOI: 10.1016/j.tibs.2016.09.001] [Citation(s) in RCA: 372] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 08/31/2016] [Accepted: 09/07/2016] [Indexed: 12/29/2022]
Abstract
In recent years, rapid discoveries have been made relating to Ca2+ handling at specific organelles that have important implications for whole-cell Ca2+ homeostasis. In particular, the structures of the endoplasmic reticulum (ER) Ca2+ channels revealed by electron cryomicroscopy (cryo-EM), continuous updates on the structure, regulation, and role of the mitochondrial calcium uniporter (MCU) complex, and the analysis of lysosomal Ca2+ signaling are milestones on the route towards a deeper comprehension of the complexity of global Ca2+ signaling. In this review we summarize recent discoveries on the regulation of interorganellar Ca2+ homeostasis and its role in pathophysiology.
Collapse
Affiliation(s)
- Anna Raffaello
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy.
| | - Cristina Mammucari
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy.
| | - Gaia Gherardi
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; Neuroscience Institute, National Research Council, 35131 Padua, Italy.
| |
Collapse
|
35
|
Bartlett PJ, Metzger W, Gaspers LD, Thomas AP. Differential Regulation of Multiple Steps in Inositol 1,4,5-Trisphosphate Signaling by Protein Kinase C Shapes Hormone-stimulated Ca2+ Oscillations. J Biol Chem 2015; 290:18519-33. [PMID: 26078455 DOI: 10.1074/jbc.m115.657767] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Indexed: 11/06/2022] Open
Abstract
How Ca(2+) oscillations are generated and fine-tuned to yield versatile downstream responses remains to be elucidated. In hepatocytes, G protein-coupled receptor-linked Ca(2+) oscillations report signal strength via frequency, whereas Ca(2+) spike amplitude and wave velocity remain constant. IP3 uncaging also triggers oscillatory Ca(2+) release, but, in contrast to hormones, Ca(2+) spike amplitude, width, and wave velocity were dependent on [IP3] and were not perturbed by phospholipase C (PLC) inhibition. These data indicate that oscillations elicited by IP3 uncaging are driven by the biphasic regulation of the IP3 receptor by Ca(2+), and, unlike hormone-dependent responses, do not require PLC. Removal of extracellular Ca(2+) did not perturb Ca(2+) oscillations elicited by IP3 uncaging, indicating that reloading of endoplasmic reticulum stores via plasma membrane Ca(2+) influx does not entrain the signal. Activation and inhibition of PKC attenuated hormone-induced Ca(2+) oscillations but had no effect on Ca(2+) increases induced by uncaging IP3. Importantly, PKC activation and inhibition differentially affected Ca(2+) spike frequencies and kinetics. PKC activation amplifies negative feedback loops at the level of G protein-coupled receptor PLC activity and/or IP3 metabolism to attenuate IP3 levels and suppress the generation of Ca(2+) oscillations. Inhibition of PKC relieves negative feedback regulation of IP3 accumulation and, thereby, shifts Ca(2+) oscillations toward sustained responses or dramatically prolonged spikes. PKC down-regulation attenuates phenylephrine-induced Ca(2+) wave velocity, whereas responses to IP3 uncaging are enhanced. The ability to assess Ca(2+) responses in the absence of PLC activity indicates that IP3 receptor modulation by PKC regulates Ca(2+) release and wave velocity.
Collapse
Affiliation(s)
- Paula J Bartlett
- From the Department of Pharmacology and Physiology, New Jersey Medical School Rutgers, The State University of New Jersey, Newark, New Jersey 07103
| | - Walson Metzger
- From the Department of Pharmacology and Physiology, New Jersey Medical School Rutgers, The State University of New Jersey, Newark, New Jersey 07103
| | - Lawrence D Gaspers
- From the Department of Pharmacology and Physiology, New Jersey Medical School Rutgers, The State University of New Jersey, Newark, New Jersey 07103
| | - Andrew P Thomas
- From the Department of Pharmacology and Physiology, New Jersey Medical School Rutgers, The State University of New Jersey, Newark, New Jersey 07103
| |
Collapse
|