1
|
Kang M, Senatore AJ, Naughton H, McTigue M, Beltman RJ, Herppich AA, Pflum MKH, Howe AK. Protein kinase A is a functional component of focal adhesions. J Biol Chem 2024; 300:107234. [PMID: 38552737 PMCID: PMC11044056 DOI: 10.1016/j.jbc.2024.107234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 03/06/2024] [Accepted: 03/17/2024] [Indexed: 04/09/2024] Open
Abstract
Focal adhesions (FAs) form the junction between extracellular matrix (ECM)-bound integrins and the actin cytoskeleton and also transmit signals that regulate cell adhesion, cytoskeletal dynamics, and cell migration. While many of these signals are rooted in reversible tyrosine phosphorylation, phosphorylation of FA proteins on Ser/Thr residues is far more abundant yet its mechanisms and consequences are far less understood. The cAMP-dependent protein kinase (protein kinase A; PKA) has important roles in cell adhesion and cell migration and is both an effector and regulator of integrin-mediated adhesion to the ECM. Importantly, subcellular localization plays a critically important role in specifying PKA function. Here, we show that PKA is present in isolated FA-cytoskeleton complexes and active within FAs in live cells. Furthermore, using kinase-catalyzed biotinylation of isolated FA-cytoskeleton complexes, we identify 53 high-stringency candidate PKA substrates within FAs. From this list, we validate tensin-3 (Tns3)-a well-established molecular scaffold, regulator of cell migration, and a component of focal and fibrillar adhesions-as a novel direct substrate for PKA. These observations identify a new pathway for phospho-regulation of Tns3 and, importantly, establish a new and important niche for localized PKA signaling and thus provide a foundation for further investigation of the role of PKA in the regulation of FA dynamics and signaling.
Collapse
Affiliation(s)
- Mingu Kang
- Department of Pharmacology, Larner College of Medicine, University of Vermont Cancer Center, Burlington, Vermont, USA; Department of Molecular Physiology & Biophysics, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Amanda J Senatore
- Department of Pharmacology, Larner College of Medicine, University of Vermont Cancer Center, Burlington, Vermont, USA; Department of Molecular Physiology & Biophysics, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Hannah Naughton
- Department of Pharmacology, Larner College of Medicine, University of Vermont Cancer Center, Burlington, Vermont, USA; Department of Molecular Physiology & Biophysics, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Madeline McTigue
- Department of Pharmacology, Larner College of Medicine, University of Vermont Cancer Center, Burlington, Vermont, USA; Department of Molecular Physiology & Biophysics, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Rachel J Beltman
- Department of Chemistry, Wayne State University, Detroit, Michigan, USA
| | - Andrew A Herppich
- Department of Chemistry, Wayne State University, Detroit, Michigan, USA
| | - Mary Kay H Pflum
- Department of Chemistry, Wayne State University, Detroit, Michigan, USA
| | - Alan K Howe
- Department of Pharmacology, Larner College of Medicine, University of Vermont Cancer Center, Burlington, Vermont, USA; Department of Molecular Physiology & Biophysics, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA.
| |
Collapse
|
2
|
Kang M, Otani Y, Guo Y, Yan J, Goult BT, Howe AK. The focal adhesion protein talin is a mechanically gated A-kinase anchoring protein. Proc Natl Acad Sci U S A 2024; 121:e2314947121. [PMID: 38513099 PMCID: PMC10990152 DOI: 10.1073/pnas.2314947121] [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: 08/29/2023] [Accepted: 02/22/2024] [Indexed: 03/23/2024] Open
Abstract
Protein kinase A (PKA) is a ubiquitous, promiscuous kinase whose activity is specified through subcellular localization mediated by A-kinase anchoring proteins (AKAPs). PKA has complex roles as both an effector and a regulator of integrin-mediated cell adhesion to extracellular matrix (ECM). Recent observations demonstrate that PKA is an active component of focal adhesions (FA), suggesting the existence of one or more FA AKAPs. Using a promiscuous biotin ligase fused to PKA type-IIα regulatory (RIIα) subunits and subcellular fractionation, we identify the archetypal FA protein talin1 as an AKAP. Talin is a large, mechanosensitive scaffold that directly links integrins to actin filaments and promotes FA assembly by recruiting additional components in a force-dependent manner. The rod region of talin1 consists of 62 α-helices bundled into 13 rod domains, R1 to R13. Direct binding assays and NMR spectroscopy identify helix41 in the R9 subdomain of talin as the PKA binding site. PKA binding to helix41 requires unfolding of the R9 domain, which requires the linker region between R9 and R10. Experiments with single molecules and in cells manipulated to alter actomyosin contractility demonstrate that the PKA-talin interaction is regulated by mechanical force across the talin molecule. Finally, talin mutations that disrupt PKA binding also decrease levels of total and phosphorylated PKA RII subunits as well as phosphorylation of VASP, a known PKA substrate, within FA. These observations identify a mechanically gated anchoring protein for PKA, a force-dependent binding partner for talin1, and a potential pathway for adhesion-associated mechanotransduction.
Collapse
Affiliation(s)
- Mingu Kang
- Department of Pharmacology, University of Vermont Larner College of Medicine, Burlington, VT05405
- Department of Molecular Physiology and Biophysics, University of Vermont Larner College of Medicine, Burlington, VT05405
- University of Vermont Cancer Center, Burlington, VT05405
| | - Yasumi Otani
- School of Biosciences, University of Kent, Canterbury, KentCT2 7NJ, United Kingdom
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, LiverpoolL69 7ZB, United Kingdom
| | - Yanyu Guo
- Department of Physics, Mechanobiology Institute, National University of Singapore, Singapore117542, Singapore
| | - Jie Yan
- Department of Physics, Mechanobiology Institute, National University of Singapore, Singapore117542, Singapore
| | - Benjamin T. Goult
- School of Biosciences, University of Kent, Canterbury, KentCT2 7NJ, United Kingdom
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, LiverpoolL69 7ZB, United Kingdom
| | - Alan K. Howe
- Department of Pharmacology, University of Vermont Larner College of Medicine, Burlington, VT05405
- Department of Molecular Physiology and Biophysics, University of Vermont Larner College of Medicine, Burlington, VT05405
- University of Vermont Cancer Center, Burlington, VT05405
| |
Collapse
|
3
|
Kang M, Senatore AJ, Naughton H, McTigue M, Beltman RJ, Herppich AA, Pflum MKH, Howe AK. Protein Kinase A is a Functional Component of Focal Adhesions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.18.553932. [PMID: 37645771 PMCID: PMC10462105 DOI: 10.1101/2023.08.18.553932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Focal adhesions (FAs) form the junction between extracellular matrix (ECM)-bound integrins and the actin cytoskeleton and also transmit signals that regulate cell adhesion, cytoskeletal dynamics, and cell migration. While many of these signals are rooted in reversible tyrosine phosphorylation, phosphorylation of FA proteins on Ser/Thr residues is far more abundant yet its mechanisms and consequences are far less understood. The cAMP-dependent protein kinase (protein kinase A; PKA) has important roles in cell adhesion and cell migration and is both an effector and regulator of integrin-mediated adhesion to the ECM. Importantly, subcellular localization plays a critically important role in specifying PKA function. Here, we show that PKA is present in isolated FA-cytoskeleton complexes and active within FAs in live cells. Furthermore, using kinase-catalyzed biotinylation of isolated FA-cytoskeleton complexes, we identify fifty-three high-stringency candidate PKA substrates within FAs. From this list, we validate tensin-3 (Tns3) - a well-established molecular scaffold, regulator of cell migration, and component of focal and fibrillar adhesions - as a novel direct substrate for PKA. These observations identify a new pathway for phospho-regulation of Tns3 and, importantly, establish a new and important niche for localized PKA signaling and thus provide a foundation for further investigation of the role of PKA in the regulation of FA dynamics and signaling.
Collapse
|
4
|
Cepkenovic B, Friedland F, Noetzel E, Maybeck V, Offenhäusser A. Single-neuron mechanical perturbation evokes calcium plateaus that excite and modulate the network. Sci Rep 2023; 13:20669. [PMID: 38001109 PMCID: PMC10673841 DOI: 10.1038/s41598-023-47090-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Mechanical stimulation is a promising means to non-invasively excite and modulate neuronal networks with a high spatial resolution. Despite the thorough characterization of the initiation mechanism, whether or how mechanical responses disperse into non-target areas remains to be discovered. Our in vitro study demonstrates that a single-neuron deformation evokes responses that propagate to about a third of the untouched neighbors. The responses develop via calcium influx through mechanosensitive channels and regeneratively propagate through the neuronal ensemble via gap junctions. Although independent of action potentials and synapses, mechanical responses reliably evoke membrane depolarizations capable of inducing action potentials both in the target and neighbors. Finally, we show that mechanical stimulation transiently potentiates the responding assembly for further inputs, as both gain and excitability are transiently increased exclusively in neurons that respond to a neighbor's mechanical stimulation. The findings indicate a biological component affecting the spatial resolution of mechanostimulation and point to a cross-talk in broad-network mechanical stimulations. Since giga-seal formation in patch-clamp produces a similar mechanical stimulus on the neuron, our findings inform which neuroscientific questions could be reliably tackled with patch-clamp and what recovery post-gigaseal formation is necessary.
Collapse
Affiliation(s)
- Bogdana Cepkenovic
- Institute of Biological Information Processing: Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße 1, 52428, Jülich, Germany
- RWTH Aachen University, Templergraben 55, 52062, Aachen, Germany
| | - Florian Friedland
- Institute of Biological Information Processing: Mechanobiology (IBI-2), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße 1, 52428, Jülich, Germany
| | - Erik Noetzel
- Institute of Biological Information Processing: Mechanobiology (IBI-2), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße 1, 52428, Jülich, Germany
| | - Vanessa Maybeck
- Institute of Biological Information Processing: Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße 1, 52428, Jülich, Germany.
| | - Andreas Offenhäusser
- Institute of Biological Information Processing: Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße 1, 52428, Jülich, Germany
- RWTH Aachen University, Templergraben 55, 52062, Aachen, Germany
| |
Collapse
|
5
|
Staehr C, Aalkjaer C, Matchkov V. The vascular Na,K-ATPase: clinical implications in stroke, migraine, and hypertension. Clin Sci (Lond) 2023; 137:1595-1618. [PMID: 37877226 PMCID: PMC10600256 DOI: 10.1042/cs20220796] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 10/26/2023]
Abstract
In the vascular wall, the Na,K-ATPase plays an important role in the control of arterial tone. Through cSrc signaling, it contributes to the modulation of Ca2+ sensitivity in vascular smooth muscle cells. This review focuses on the potential implication of Na,K-ATPase-dependent intracellular signaling pathways in severe vascular disorders; ischemic stroke, familial migraine, and arterial hypertension. We propose similarity in the detrimental Na,K-ATPase-dependent signaling seen in these pathological conditions. The review includes a retrospective proteomics analysis investigating temporal changes after ischemic stroke. The analysis revealed that the expression of Na,K-ATPase α isoforms is down-regulated in the days and weeks following reperfusion, while downstream Na,K-ATPase-dependent cSrc kinase is up-regulated. These results are important since previous studies have linked the Na,K-ATPase-dependent cSrc signaling to futile recanalization and vasospasm after stroke. The review also explores a link between the Na,K-ATPase and migraine with aura, as reduced expression or pharmacological inhibition of the Na,K-ATPase leads to cSrc kinase signaling up-regulation and cerebral hypoperfusion. The review discusses the role of an endogenous cardiotonic steroid-like compound, ouabain, which binds to the Na,K-ATPase and initiates the intracellular cSrc signaling, in the pathophysiology of arterial hypertension. Currently, our understanding of the precise control mechanisms governing the Na,K-ATPase/cSrc kinase regulation in the vascular wall is limited. Understanding the role of vascular Na,K-ATPase signaling is essential for developing targeted treatments for cerebrovascular disorders and hypertension, as the Na,K-ATPase is implicated in the pathogenesis of these conditions and may contribute to their comorbidity.
Collapse
Affiliation(s)
- Christian Staehr
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000 Aarhus, Denmark
- Department of Renal Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 35, Aarhus, Denmark
| | - Christian Aalkjaer
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000 Aarhus, Denmark
- Danish Cardiovascular Academy, Høegh-Guldbergsgade 10, 8000 Aarhus, Denmark
| | - Vladimir V. Matchkov
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000 Aarhus, Denmark
| |
Collapse
|
6
|
Kang M, Otani Y, Guo Y, Yan J, Goult BT, Howe AK. The focal adhesion protein talin is a mechanically-gated A-kinase anchoring protein (AKAP). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.20.554038. [PMID: 37645895 PMCID: PMC10462126 DOI: 10.1101/2023.08.20.554038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
The cAMP-dependent protein kinase (Protein Kinase A; PKA) is a ubiquitous, promiscuous kinase whose activity is focused and specified through subcellular localization mediated by A-kinase anchoring proteins (AKAPs). PKA has complex roles as both an effector and a regulator of integrin-mediated cell adhesion to the extracellular matrix (ECM). Recent observations demonstrate that PKA is an active component of focal adhesions (FA), intracellular complexes coupling ECM-bound integrins to the actin cytoskeleton, suggesting the existence of one or more FA AKAPs. Using a combination of a promiscuous biotin ligase fused to PKA type-IIα regulatory (RIIα) subunits and subcellular fractionation, we identify the archetypal FA protein talin1 as an AKAP. Talin is a large, mechanosensitive scaffold that directly links integrins to actin filaments and promotes FA assembly by recruiting additional components in a force-dependent manner. The rod region of talin1 consists of 62 α-helices bundled into 13 rod domains, R1-R13. Direct binding assays and nuclear magnetic resonance spectroscopy identify helix41 in the R9 subdomain of talin as the PKA binding site. PKA binding to helix41 requires unfolding of the R9 domain, which requires the linker region between R9 and R10. Finally, single-molecule experiments with talin1 and PKA, and experiments in cells manipulated to alter actomyosin contractility demonstrate that the PKA-talin interaction is regulated by mechanical force across the talin molecule. These observations identify the first mechanically-gated anchoring protein for PKA, a new force-dependent binding partner for talin1, and thus a new mechanism for coupling cellular tension and signal transduction.
Collapse
|
7
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|
8
|
Martín-Aragón Baudel M, Hong J, Hell JW, Nieves-Cintrón M, Navedo MF. Mechanisms of Vascular Ca V1.2 Channel Regulation During Diabetic Hyperglycemia. Handb Exp Pharmacol 2023; 279:41-58. [PMID: 36598607 DOI: 10.1007/164_2022_628] [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: 01/05/2023]
Abstract
Diabetes is a leading cause of disability and mortality worldwide. A major underlying factor in diabetes is the excessive glucose levels in the bloodstream (e.g., hyperglycemia). Vascular complications directly result from this metabolic abnormality, leading to disabling and life-threatening conditions. Dysfunction of vascular smooth muscle cells is a well-recognized factor mediating vascular complications during diabetic hyperglycemia. The function of vascular smooth muscle cells is exquisitely controlled by different ion channels. Among the ion channels, the L-type CaV1.2 channel plays a key role as it is the main Ca2+ entry pathway regulating vascular smooth muscle contractile state. The activity of CaV1.2 channels in vascular smooth muscle is altered by diabetic hyperglycemia, which may contribute to vascular complications. In this chapter, we summarize the current understanding of the regulation of CaV1.2 channels in vascular smooth muscle by different signaling pathways. We place special attention on the regulation of CaV1.2 channel activity in vascular smooth muscle by a newly uncovered AKAP5/P2Y11/AC5/PKA/CaV1.2 axis that is engaged during diabetic hyperglycemia. We further describe the pathophysiological implications of activation of this axis as it relates to myogenic tone and vascular reactivity and propose that this complex may be targeted for developing therapies to treat diabetic vascular complications.
Collapse
Affiliation(s)
| | - Junyoung Hong
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - Johannes W Hell
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | | | - Manuel F Navedo
- Department of Pharmacology, University of California Davis, Davis, CA, USA.
| |
Collapse
|
9
|
Hu XQ, Zhang L. Oxidative Regulation of Vascular Ca v1.2 Channels Triggers Vascular Dysfunction in Hypertension-Related Disorders. Antioxidants (Basel) 2022; 11:antiox11122432. [PMID: 36552639 PMCID: PMC9774363 DOI: 10.3390/antiox11122432] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/28/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Blood pressure is determined by cardiac output and peripheral vascular resistance. The L-type voltage-gated Ca2+ (Cav1.2) channel in small arteries and arterioles plays an essential role in regulating Ca2+ influx, vascular resistance, and blood pressure. Hypertension and preeclampsia are characterized by high blood pressure. In addition, diabetes has a high prevalence of hypertension. The etiology of these disorders remains elusive, involving the complex interplay of environmental and genetic factors. Common to these disorders are oxidative stress and vascular dysfunction. Reactive oxygen species (ROS) derived from NADPH oxidases (NOXs) and mitochondria are primary sources of vascular oxidative stress, whereas dysfunction of the Cav1.2 channel confers increased vascular resistance in hypertension. This review will discuss the importance of ROS derived from NOXs and mitochondria in regulating vascular Cav1.2 and potential roles of ROS-mediated Cav1.2 dysfunction in aberrant vascular function in hypertension, diabetes, and preeclampsia.
Collapse
|
10
|
Forzisi E, Sesti F. Non-conducting functions of ion channels: The case of integrin-ion channel complexes. Channels (Austin) 2022; 16:185-197. [PMID: 35942524 PMCID: PMC9364710 DOI: 10.1080/19336950.2022.2108565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Started as an academic curiosity more than two decades ago, the idea that ion channels can regulate cellular processes in ways that do not depend on their conducting properties (non-ionic functions) gained traction and is now a flourishing area of research. Channels can regulate physiological processes including actin cytoskeletal remodeling, cell motility, excitation-contraction coupling, non-associative learning and embryogenesis, just to mention some, through non-ionic functions. When defective, non-ionic functions can give rise to channelopathies involved in cancer, neurodegenerative disease and brain trauma. Ion channels exert their non-ionic functions through a variety of mechanisms that range from physical coupling with other proteins, to possessing enzymatic activity, to assembling with signaling molecules. In this article, we take stock of the field and review recent findings. The concept that emerges, is that one of the most common ways through which channels acquire non-ionic attributes, is by assembling with integrins. These integrin-channel complexes exhibit broad genotypic and phenotypic heterogeneity and reveal a pleiotropic nature, as they appear to be capable of influencing both physiological and pathological processes.
Collapse
Affiliation(s)
- Elena Forzisi
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, NJ, USA
| | - Federico Sesti
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, NJ, USA
| |
Collapse
|
11
|
Modulation of L-type calcium channels in Alzheimer's disease: A potential therapeutic target. Comput Struct Biotechnol J 2022; 21:11-20. [PMID: 36514335 PMCID: PMC9719069 DOI: 10.1016/j.csbj.2022.11.049] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 11/28/2022] Open
Abstract
Calcium plays a fundamental role in various signaling pathways and cellular processes in the human organism. In the nervous system, voltage-gated calcium channels such as L-type calcium channels (LTCCs) are critical elements in mediating neurotransmitter release, synaptic integration and plasticity. Dysfunction of LTCCs has been implicated in both aging and Alzheimer's Disease (AD), constituting a key component of calcium hypothesis of AD. As such, LTCCs are a promising drug target in AD. However, due to their structural and functional complexity, the mechanisms by which LTCCs contribute to AD are still unclear. In this review, we briefly summarize the structure, function, and modulation of LTCCs that are the backbone for understanding pathological processes involving LTCCs. We suggest targeting molecular pathways up-regulating LTCCs in AD may be a more promising approach, given the diverse physiological functions of LTCCs and the ineffectiveness of LTCC blockers in clinical studies.
Collapse
Key Words
- AC, adenylyl cyclase
- AD, Alzheimer’s Disease
- AHP, afterhyperpolarization
- AR, adrenoceptor
- Aging
- Alzheimer’s disease
- Aβ, β-amyloid
- BIN1, bridging integrator 1
- BTZs, benzothiazepines
- CDF, calcium-dependent facilitation
- CDI, calcium-dependent inactivation
- CaMKII, calmodulin-dependent protein kinase II
- DHP, dihydropyridine
- L-type calcium channel
- LTCC, L-type calcium channels
- LTD, long-term depression
- LTP, long-term potentiation
- NFT, neurofibrillary tangles
- NMDAR, N-methyl-D-aspartate receptor
- PAA, phenylalkylamines
- PKA, protein kinase A
- PKC, protein kinase C
- PKG, protein kinase G
- SFK, Src family kinase
- Tau
- VSD, voltage sensing domain
- β-Amyloid
Collapse
|
12
|
Li Y, Yang H, He T, Zhang L, Liu C. Post-Translational Modification of Cav1.2 and its Role in Neurodegenerative Diseases. Front Pharmacol 2022; 12:775087. [PMID: 35111050 PMCID: PMC8802068 DOI: 10.3389/fphar.2021.775087] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/08/2021] [Indexed: 11/26/2022] Open
Abstract
Cav1.2 plays an essential role in learning and memory, drug addiction, and neuronal development. Intracellular calcium homeostasis is disrupted in neurodegenerative diseases because of abnormal Cav1.2 channel activity and modification of downstream Ca2+ signaling pathways. Multiple post-translational modifications of Cav1.2 have been observed and seem to be closely related to the pathogenesis of neurodegenerative diseases. The specific molecular mechanisms by which Cav1.2 channel activity is regulated remain incompletely understood. Dihydropyridines (DHPs), which are commonly used for hypertension and myocardial ischemia, have been repurposed to treat PD and AD and show protective effects. However, further studies are needed to improve delivery strategies and drug selectivity. Better knowledge of channel modulation and more specific methods for altering Cav1.2 channel function may lead to better therapeutic strategies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Yun Li
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Hong Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Tianhan He
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Liang Zhang
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chao Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
13
|
Orzeł-Gajowik K, Milewski K, Zielińska M. Insight into microRNAs-Mediated Communication between Liver and Brain: A Possible Approach for Understanding Acute Liver Failure? Int J Mol Sci 2021; 23:224. [PMID: 35008650 PMCID: PMC8745738 DOI: 10.3390/ijms23010224] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 01/11/2023] Open
Abstract
Acute liver failure (ALF) is a life-threatening consequence of hepatic function rapid loss without preexisting liver disease. ALF may result in a spectrum of neuropsychiatric symptoms that encompasses cognitive impairment, coma, and often death, collectively defined as acute hepatic encephalopathy. Micro RNAs are small non-coding RNAs that modulate gene expression and are extensively verified as biomarker candidates in various diseases. Our systematic literature review based on the last decade's reports involving a total of 852 ALF patients, determined 205 altered circulating miRNAs, of which 25 miRNAs were altered in the blood, regardless of study design and methodology. Selected 25 miRNAs, emerging predominantly from the analyses of samples obtained from acetaminophen overdosed patients, represent the most promising biomarker candidates for a diagnostic panel for symptomatic ALF. We discussed the role of selected miRNAs in the context of tissue-specific origin and its possible regulatory role for molecular pathways involved in blood-brain barrier function. The defined several common pathways for 15 differently altered miRNAs were relevant to cellular community processes, indicating loss of intercellular, structural, and functional components, which may result in blood-brain barrier impairment and brain dysfunction. However, a causational relationship between circulating miRNAs differential expression, and particular clinical features of ALF, has to be demonstrated in a further study.
Collapse
Affiliation(s)
| | | | - Magdalena Zielińska
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106 Warsaw, Poland; (K.O.-G.); (K.M.)
| |
Collapse
|
14
|
Zhang YL, Zhang RG, Chen FY, Qiu ZE, Chen L, Huang ZX, Huang J, Zhu YX, Zhao L, Zhou WL. Cellular Mechanism Underlying the Facilitation of Contractile Response Induced by Tumor Necrosis Factor-α in Mouse Tracheal Smooth Muscle. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 192:104-111. [PMID: 34756873 DOI: 10.1016/j.ajpath.2021.10.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 01/10/2023]
Abstract
The proinflammatory cytokine tumor necrosis factor-α (TNF-α) augments intracellular Ca2+ signaling and contractile responses of airway smooth muscles, leading to airway hyperresponsiveness. However, the underlying mechanism has not been fully elucidated. This study aimed to investigate the cellular mechanism of the potentiated contraction of mouse tracheal smooth muscle induced by TNF-α. The results showed that TNF-α triggered facilitation of mouse tracheal smooth muscle contraction in an epithelium-independent manner. The TNF-α-induced hypercontractility could be suppressed by the protein kinase C inhibitor GF109203X, the tyrosine kinase inhibitor genistein, the Src inhibitor PP2, or the L-type voltage-dependent Ca2+ channel blocker nifedipine. After TNF-α incubation, the α1C L-type Ca2+ channel (CaV1.2) was up-regulated in primary cultured mouse tracheal smooth muscle cells. Pronounced phosphotyrosine levels also were observed in mouse tracheas. In conclusion, this study showed that TNF-α enhanced airway smooth muscle contraction via protein kinase C-Src-CaV1.2 pathways, which provides novel insights into the pathologic role of proinflammatory cytokines in mediating airway hyperresponsiveness.
Collapse
Affiliation(s)
- Yi-Lin Zhang
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Rui-Gang Zhang
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China; Department of Physiology, Basic Medical School, Guangdong Medical University, Zhanjiang, China
| | - Feng-Ying Chen
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; Department of Pathology, The Maternal and Child Health Care Hospital of HuaDu District (Huzhong Hospital), Guangzhou, China
| | - Zhuo-Er Qiu
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Lei Chen
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ze-Xin Huang
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jiehong Huang
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yun-Xin Zhu
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Lei Zhao
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China.
| | - Wen-Liang Zhou
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
15
|
Simons P, Rinaldi DA, Bondu V, Kell AM, Bradfute S, Lidke D, Buranda T. Integrin activation is an essential component of SARS-CoV-2 infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 34312625 DOI: 10.1101/2021.07.20.453118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cellular entry of coronaviruses depends on binding of the viral spike (S) protein to a specific cellular receptor, the angiotensin-converting enzyme 2 (ACE2). Furthermore, the viral spike protein expresses an RGD motif, suggesting that cell surface integrins may be attachment co-receptors. However, using infectious SARS-CoV-2 requires a biosafety level 3 laboratory (BSL-3), which limits the techniques that can be used to study the mechanism of cell entry. Here, we UV-inactivated SARS-CoV-2 and fluorescently labeled the envelope membrane with octadecyl rhodamine B (R18) to explore the role of integrin activation in mediating both cell entry and productive infection. We used flow cytometry and confocal fluorescence microscopy to show that fluorescently labeled SARS-CoV-2 R18 particles engage basal-state integrins. Furthermore, we demonstrate that Mn 2+ , which activates integrins and induces integrin extension, enhances cell binding and entry of SARS-CoV-2 R18 in proportion to the fraction of integrins activated. We also show that one class of integrin antagonist, which binds to the αI MIDAS site and stabilizes the inactive, closed conformation, selectively inhibits the engagement of SARS-CoV-2 R18 with basal state integrins, but is ineffective against Mn 2+ -activated integrins. At the same time, RGD-integrin antagonists inhibited SARS-CoV-2 R18 binding regardless of integrin activity state. Integrins transmit signals bidirectionally: 'inside-out' signaling primes the ligand binding function of integrins via a talin dependent mechanism and 'outside-in' signaling occurs downstream of integrin binding to macromolecular ligands. Outside-in signaling is mediated by Gα 13 and induces cell spreading, retraction, migration, and proliferation. Using cell-permeable peptide inhibitors of talin, and Gα 13 binding to the cytoplasmic tail of an integrin's β subunit, we further demonstrate that talin-mediated signaling is essential for productive infection by SARS-CoV-2.
Collapse
|
16
|
Calcium in Cell-Extracellular Matrix Interactions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:1079-1102. [PMID: 31646546 DOI: 10.1007/978-3-030-12457-1_43] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In multicellular organisms, the cells are surrounded by persistent, dynamic extracellular matrix (ECM), the largest calcium reservoir in animals. ECM regulates several aspects of cell behavior including cell migration and adhesion, survival, gene expression and differentiation, thus playing a significant role in health and disease. Calcium is reported to be important in the assembly of ECM, where it binds to many ECM proteins. While serving as a calcium reservoir, ECM macromolecules can directly interact with cell surface receptors resulting in calcium transport across the membrane. This chapter mainly focusses on the role of cell-ECM interactions in cellular calcium regulation and how calcium itself mediates these interactions.
Collapse
|
17
|
Knock GA. NADPH oxidase in the vasculature: Expression, regulation and signalling pathways; role in normal cardiovascular physiology and its dysregulation in hypertension. Free Radic Biol Med 2019; 145:385-427. [PMID: 31585207 DOI: 10.1016/j.freeradbiomed.2019.09.029] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/29/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023]
Abstract
The last 20-25 years have seen an explosion of interest in the role of NADPH oxidase (NOX) in cardiovascular function and disease. In vascular smooth muscle and endothelium, NOX generates reactive oxygen species (ROS) that act as second messengers, contributing to the control of normal vascular function. NOX activity is altered in response to a variety of stimuli, including G-protein coupled receptor agonists, growth-factors, perfusion pressure, flow and hypoxia. NOX-derived ROS are involved in smooth muscle constriction, endothelium-dependent relaxation and smooth muscle growth, proliferation and migration, thus contributing to the fine-tuning of blood flow, arterial wall thickness and vascular resistance. Through reversible oxidative modification of target proteins, ROS regulate the activity of protein tyrosine phosphatases, kinases, G proteins, ion channels, cytoskeletal proteins and transcription factors. There is now considerable, but somewhat contradictory evidence that NOX contributes to the pathogenesis of hypertension through oxidative stress. Specific NOX isoforms have been implicated in endothelial dysfunction, hyper-contractility and vascular remodelling in various animal models of hypertension, pulmonary hypertension and pulmonary arterial hypertension, but also have potential protective effects, particularly NOX4. This review explores the multiplicity of NOX function in the healthy vasculature and the evidence for and against targeting NOX for antihypertensive therapy.
Collapse
Affiliation(s)
- Greg A Knock
- Dpt. of Inflammation Biology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, UK.
| |
Collapse
|
18
|
McKenzie AJ, Svec KV, Williams TF, Howe AK. Protein kinase A activity is regulated by actomyosin contractility during cell migration and is required for durotaxis. Mol Biol Cell 2019; 31:45-58. [PMID: 31721649 PMCID: PMC6938270 DOI: 10.1091/mbc.e19-03-0131] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Dynamic subcellular regulation of protein kinase A (PKA) activity is important for the motile behavior of many cell types, yet the mechanisms governing PKA activity during cell migration remain largely unknown. The motility of SKOV-3 epithelial ovarian cancer (EOC) cells has been shown to be dependent both on localized PKA activity and, more recently, on mechanical reciprocity between cellular tension and extracellular matrix rigidity. Here, we investigated the possibility that PKA is regulated by mechanical signaling during migration. We find that localized PKA activity in migrating cells rapidly decreases upon inhibition of actomyosin contractility (specifically, of myosin ATPase, Rho kinase, or myosin light-chain kinase activity). Moreover, PKA activity is spatially and temporally correlated with cellular traction forces in migrating cells. Additionally, PKA is rapidly and locally activated by mechanical stretch in an actomyosin contractility-dependent manner. Finally, inhibition of PKA activity inhibits mechanically guided migration, also known as durotaxis. These observations establish PKA as a locally regulated effector of cellular mechanotransduction and as a regulator of mechanically guided cell migration.
Collapse
Affiliation(s)
- Andrew J McKenzie
- Department of Pharmacology.,University of Vermont Cancer Center, and
| | - Kathryn V Svec
- Department of Pharmacology.,University of Vermont Cancer Center, and
| | - Tamara F Williams
- Department of Pharmacology.,University of Vermont Cancer Center, and
| | - Alan K Howe
- Department of Pharmacology.,University of Vermont Cancer Center, and.,Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT 05405
| |
Collapse
|
19
|
Liu W, Hashimoto T, Yamashita T, Hirano K. Coagulation factor XI induces Ca 2+ response and accelerates cell migration in vascular smooth muscle cells via proteinase-activated receptor 1. Am J Physiol Cell Physiol 2019; 316:C377-C392. [PMID: 30566391 DOI: 10.1152/ajpcell.00426.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Activated coagulation factor XI (FXIa) is a serine proteinase that plays a key role in the intrinsic coagulation pathway. The analysis of FXI-knockout mice has indicated the contribution of FXI to the pathogenesis of atherosclerosis. However, the underlying mechanism remains unknown. We hypothesized that FXIa exerts vascular smooth muscle effects via proteinase-activated receptor 1 (PAR1). Fura-2 fluorometry revealed that FXIa elicited intracellular Ca2+ signal in rat embryo aorta smooth muscle A7r5 cells. The influx of extracellular Ca2+ played a greater role in generating Ca2+ signal than the Ca2+ release from intracellular stores. The FXIa-induced Ca2+ signal was abolished by the pretreatment with atopaxar, an antagonist of PAR1, or 4-amidinophenylmethanesulfonyl fluoride (p-APMSF), an inhibitor of proteinase, while it was also lost in embryonic fibroblasts derived from PAR1-/- mice. FXIa cleaved the recombinant protein containing the extracellular region of PAR1 at the same site (R45/S46) as that of thrombin, a canonical PAR1 agonist. The FXIa-induced Ca2+ influx was inhibited by diltiazem, an L-type Ca2+ channel blocker, and by siRNA targeted to CaV1.2. The FXIa-induced Ca2+ influx was also inhibited by GF109203X and rottlerin, inhibitors of protein kinase C. In a wound healing assay, FXIa increased the rate of cell migration by 2.46-fold of control, which was partly inhibited by atopaxar or diltiazem. In conclusion, FXIa mainly elicits the Ca2+ signal via the PAR1/CaV1.2-mediated Ca2+ influx and accelerates the migration in vascular smooth muscle cells. The present study provides the first evidence that FXIa exerts a direct cellular effect on vascular smooth muscle.
Collapse
Affiliation(s)
- Wenhua Liu
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University , Kagawa , Japan
| | - Takeshi Hashimoto
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University , Kagawa , Japan
| | - Tetsuo Yamashita
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University , Kagawa , Japan
| | - Katsuya Hirano
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University , Kagawa , Japan
| |
Collapse
|
20
|
Abstract
The Na,K-ATPase is an enzyme essential for ion homeostasis in all cells. Over the last decades, it has been well-established that in addition to the transport of Na+/K+ over the cell membrane, the Na,K-ATPase acts as a receptor transducing humoral signals intracellularly. It has been suggested that ouabain-like compounds serve as endogenous modulators of this Na,K-ATPase signal transduction. The molecular mechanisms underlying Na,K-ATPase signaling are complicated and suggest the confluence of divergent biological pathways. This review discusses recent updates on the Na,K-ATPase signaling pathways characterized or suggested in vascular smooth muscle cells. The conventional view on this signaling is based on a microdomain structure where the Na,K-ATPase controls the Na,Ca-exchanger activity via modulation of intracellular Na+ in the spatially restricted submembrane space. This, in turn, affects intracellular Ca2+ and Ca2+ load in the sarcoplasmic reticulum leading to modulation of contractility as well as gene expression. An ion-transport-independent signal transduction from the Na,K-ATPase is based on molecular interactions. This was primarily characterized in other cell types but recently also demonstrated in vascular smooth muscles. The downstream signaling from the Na,K-ATPase includes Src and phosphatidylinositol-4,5-bisphosphate 3 kinase signaling pathways and generation of reactive oxygen species. Moreover, in vascular smooth muscle cells the interaction between the Na,K-ATPase and proteins responsible for Ca2+ homeostasis, e.g., phospholipase C and inositol triphosphate receptors, contributes to an integration of the signaling pathways. Recent update on the Na,K-ATPase dependent intracellular signaling and the significance for physiological functions and pathophysiological changes are discussed in this review.
Collapse
|
21
|
Bouzinova EV, Hangaard L, Staehr C, Mazur A, Ferreira A, Chibalin AV, Sandow SL, Xie Z, Aalkjaer C, Matchkov VV. The α2 isoform Na,K-ATPase modulates contraction of rat mesenteric small artery via cSrc-dependent Ca 2+ sensitization. Acta Physiol (Oxf) 2018; 224:e13059. [PMID: 29480968 DOI: 10.1111/apha.13059] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 02/06/2018] [Accepted: 02/16/2018] [Indexed: 12/11/2022]
Abstract
AIMS The Na,K-ATPase is involved in a large number of regulatory activities including cSrc-dependent signalling. Upon inhibition of the Na,K-ATPase with ouabain, cSrc activation is shown to occur in many cell types. This study tests the hypothesis that acute potentiation of agonist-induced contraction by ouabain is mediated through Na,K-ATPase-cSrc signalling-dependent sensitization of vascular smooth muscle cells to Ca2+ . METHODS Agonist-induced rat mesenteric small artery contraction was examined in vitro under isometric conditions and in vivo in anaesthetized rats. Arterial wall tension and [Ca2+ ]i in vascular smooth muscle cells were measured simultaneously. Changes in cSrc and myosin phosphatase targeting protein 1 (MYPT1) phosphorylation were analysed by Western blot. Protein expression was examined with immunohistochemistry. The α1 and α2 isoforms of the Na,K-ATPase were transiently downregulated by siRNA transfection in vivo. RESULTS Ten micromolar ouabain, but not digoxin, potentiated contraction to noradrenaline. This effect was not endothelium-dependent. Ouabain sensitized smooth muscle cells to Ca2+ , and this was associated with increased phosphorylation of cSrc and MYPT1. Inhibition of tyrosine kinase by genistein, PP2 or pNaKtide abolished the potentiating effect of ouabain on arterial contraction and Ca2+ sensitization. Downregulation of the Na,K-ATPase α2 isoform made arterial contraction insensitive to ouabain and tyrosine kinase inhibition. CONCLUSION Data suggest that micromolar ouabain potentiates agonist-induced contraction of rat mesenteric small artery via Na,K-ATPase-dependent cSrc activation, which increases Ca2+ sensitization of vascular smooth muscle cells by MYPT1 phosphorylation. This mechanism may be critical for acute control of vascular tone.
Collapse
Affiliation(s)
- E. V. Bouzinova
- Department of Biomedicine; Aarhus University; Aarhus C Denmark
| | - L. Hangaard
- Department of Biomedicine; Aarhus University; Aarhus C Denmark
| | - C. Staehr
- Department of Biomedicine; Aarhus University; Aarhus C Denmark
| | - A. Mazur
- Department of Biomedicine; Aarhus University; Aarhus C Denmark
| | - A. Ferreira
- Department of Biomedicine; Aarhus University; Aarhus C Denmark
| | - A. V. Chibalin
- Department of Molecular Medicine and Surgery; Integrative Physiology; Karolinska Institutet; Stockholm Sweden
| | - S. L. Sandow
- Faculty of Science, Health, Education and Engineering; University of the Sunshine Coast; Maroochydore Qld Australia
| | - Z. Xie
- Marshall Institute for Interdisciplinary Research; Marshall University; Huntington WV USA
| | - C. Aalkjaer
- Department of Biomedicine; Aarhus University; Aarhus C Denmark
| | - V. V. Matchkov
- Department of Biomedicine; Aarhus University; Aarhus C Denmark
| |
Collapse
|
22
|
Yip KP, Balasubramanian L, Kan C, Wang L, Liu R, Ribeiro-Silva L, Sham JSK. Intraluminal pressure triggers myogenic response via activation of calcium spark and calcium-activated chloride channel in rat renal afferent arteriole. Am J Physiol Renal Physiol 2018; 315:F1592-F1600. [PMID: 30089032 DOI: 10.1152/ajprenal.00239.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Myogenic contraction of renal arterioles is an important regulatory mechanism for renal blood flow autoregulation. We have previously demonstrated that integrin-mediated mechanical force increases the occurrence of Ca2+ sparks in freshly isolated renal vascular smooth muscle cells (VSMCs). To further test whether the generation of Ca2+ sparks is a downstream signal of mechanotransduction in pressure-induced myogenic constriction, the relationship between Ca2+ sparks and transmural perfusion pressure was investigated in intact VSMCs of pressurized rat afferent arterioles. Spontaneous Ca2+ sparks were found in VSMCs when afferent arterioles were perfused at 80 mmHg. The spark frequency was significantly increased when perfusion pressure was increased to 120 mmHg. A similar increase of spark frequency was also observed in arterioles stimulated with β1-integrin-activating antibody. Moreover, spark frequency was significantly higher in arterioles of spontaneous hypertensive rats at 80 and 120 mmHg. Spontaneous membrane current recorded using whole cell perforated patch in renal VSMCs showed predominant activity of spontaneous transient inward currents instead of spontaneous transient outward currents when holding potential was set close to physiological resting membrane potential. Real-time PCR and immunohistochemistry confirmed the expression of Ca2+-activated Cl- channel (ClCa) TMEM16A in renal VSMCs. Inhibition of TMEM16A with T16Ainh-A01 impaired the pressure-induced myogenic contraction in perfused afferent arterioles. Our study, for the first time to our knowledge, detected Ca2+ sparks in VSMCs of intact afferent arterioles, and their frequencies were positively modulated by the perfusion pressure. Our results suggest that Ca2+ sparks may couple to ClCa channels and trigger pressure-induced myogenic constriction via membrane depolarization.
Collapse
Affiliation(s)
- Kay-Pong Yip
- Department of Molecular Pharmacology and Physiology, University of South Florida , Tampa, Florida
| | - Lavanya Balasubramanian
- Department of Molecular Pharmacology and Physiology, University of South Florida , Tampa, Florida
| | - Chen Kan
- Department of Industrial, Manufacturing, and System Engineering, University of Texas at Arlington , Arlington, Texas
| | - Lei Wang
- Department of Molecular Pharmacology and Physiology, University of South Florida , Tampa, Florida
| | - Ruisheng Liu
- Department of Molecular Pharmacology and Physiology, University of South Florida , Tampa, Florida
| | - Luisa Ribeiro-Silva
- Department of Molecular Pharmacology and Physiology, University of South Florida , Tampa, Florida
| | - James S K Sham
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine , Baltimore, Maryland
| |
Collapse
|
23
|
Maffioli E, Schulte C, Nonnis S, Grassi Scalvini F, Piazzoni C, Lenardi C, Negri A, Milani P, Tedeschi G. Proteomic Dissection of Nanotopography-Sensitive Mechanotransductive Signaling Hubs that Foster Neuronal Differentiation in PC12 Cells. Front Cell Neurosci 2018; 11:417. [PMID: 29354032 PMCID: PMC5758595 DOI: 10.3389/fncel.2017.00417] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/12/2017] [Indexed: 12/11/2022] Open
Abstract
Neuronal cells are competent in precisely sensing nanotopographical features of their microenvironment. The perceived microenvironmental information will be “interpreted” by mechanotransductive processes and impacts on neuronal functioning and differentiation. Attempts to influence neuronal differentiation by engineering substrates that mimic appropriate extracellular matrix (ECM) topographies are hampered by the fact that profound details of mechanosensing/-transduction complexity remain elusive. Introducing omics methods into these biomaterial approaches has the potential to provide a deeper insight into the molecular processes and signaling cascades underlying mechanosensing/-transduction but their exigence in cellular material is often opposed by technical limitations of major substrate top-down fabrication methods. Supersonic cluster beam deposition (SCBD) allows instead the bottom-up fabrication of nanostructured substrates over large areas characterized by a quantitatively controllable ECM-like nanoroughness that has been recently shown to foster neuron differentiation and maturation. Exploiting this capacity of SCBD, we challenged mechanosensing/-transduction and differentiative behavior of neuron-like PC12 cells with diverse nanotopographies and/or changes of their biomechanical status, and analyzed their phosphoproteomic profiles in these settings. Versatile proteins that can be associated to significant processes along the mechanotransductive signal sequence, i.e., cell/cell interaction, glycocalyx and ECM, membrane/f-actin linkage and integrin activation, cell/substrate interaction, integrin adhesion complex, actomyosin organization/cellular mechanics, nuclear organization, and transcriptional regulation, were affected. The phosphoproteomic data suggested furthermore an involvement of ILK, mTOR, Wnt, and calcium signaling in these nanotopography- and/or cell mechanics-related processes. Altogether, potential nanotopography-sensitive mechanotransductive signaling hubs participating in neuronal differentiation were dissected.
Collapse
Affiliation(s)
- Elisa Maffioli
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy
| | - Carsten Schulte
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Simona Nonnis
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Francesca Grassi Scalvini
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Claudio Piazzoni
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy
| | - Cristina Lenardi
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy
| | - Armando Negri
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Paolo Milani
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy
| | - Gabriella Tedeschi
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| |
Collapse
|
24
|
Li JT, Wang WQ, Wang L, Liu NN, Zhao YL, Zhu XS, Liu QQ, Gao CF, Yang AG, Jia LT. Subanesthetic isoflurane relieves zymosan-induced neutrophil inflammatory response by targeting NMDA glutamate receptor and Toll-like receptor 2 signaling. Oncotarget 2017; 7:31772-89. [PMID: 27144523 PMCID: PMC5077975 DOI: 10.18632/oncotarget.9091] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 04/18/2016] [Indexed: 01/13/2023] Open
Abstract
Neutrophil release of NO/ONOO− induces endothelial cell barrier dysfunction in inflammatory acute lung injury (ALI). Previous studies using zymosan-triggered inflammation and ALI model revealed that zymosan promotes inducible NO synthase (iNOS) expression in neutrophils, and that isoflurane inhibits zymosan-induced oxidative stress and iNOS biosynthesis. However, the underlying mechanisms remain largely unknown. We found here that in zymosan-primed neutrophils, iNOS is transcriptionally activated by NF-κB, whose nuclear translocation is triggered by excessive reactive oxygen species (ROS) and consequently activated p38 MAPK. ROS production is attributed to zymosan-initiated Toll-like receptor 2 (TLR2) signaling, in which the adaptor MyD88 recruits and activates c-Src, and c-Src activates NADPH oxidase to generate ROS. Subanesthetic isoflurane counteracts the aforementioned zymosan-induced signaling by targeting N-methyl-D-aspartic acid (NMDA) glutamate receptor and thereby suppressing calcium influx and c-Src activation. Whereas iNOS accelerates NO/ONOO− production in neutrophils which eventually promote protein leak from pulmonary microvascular endothelial cells (PMVEC), isoflurane reduced NO/ONOO− release from zymosan-treated neutrophils, and thus relieves trans- PMVEC protein leak. This study provides novel insights into the roles of neutrophils and the underlying mechanisms in zymosan-induced ALI, and has implications for the therapeutic potential of subanesthetic isoflurane in attenuating inflammatory responses causing lung endothelial cell damage.
Collapse
Affiliation(s)
- Jun-Tang Li
- Centre of Inflammation and Cancer Research, 150th Central Hospital of PLA, Luoyang, Henan, China.,State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China.,State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wei-Qi Wang
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | | | - Ning-Ning Liu
- Centre of Inflammation and Cancer Research, 150th Central Hospital of PLA, Luoyang, Henan, China
| | - Ya-Li Zhao
- Centre of Inflammation and Cancer Research, 150th Central Hospital of PLA, Luoyang, Henan, China
| | - Xiao-Shan Zhu
- Centre of Inflammation and Cancer Research, 150th Central Hospital of PLA, Luoyang, Henan, China
| | - Qin-Qin Liu
- Centre of Inflammation and Cancer Research, 150th Central Hospital of PLA, Luoyang, Henan, China
| | - Chun-Fang Gao
- Centre of Inflammation and Cancer Research, 150th Central Hospital of PLA, Luoyang, Henan, China
| | - An-Gang Yang
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lin-Tao Jia
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
25
|
Wu X, Muthuchamy M, Reddy DS. Atomic force microscopy investigations of fibronectin and α5β1-integrin signaling in neuroplasticity and seizure susceptibility in experimental epilepsy. Epilepsy Res 2017; 138:71-80. [DOI: 10.1016/j.eplepsyres.2017.10.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 09/15/2017] [Accepted: 10/19/2017] [Indexed: 12/15/2022]
|
26
|
Kashihara T, Nakada T, Kojima K, Takeshita T, Yamada M. Angiotensin II activates Ca V 1.2 Ca 2+ channels through β-arrestin2 and casein kinase 2 in mouse immature cardiomyocytes. J Physiol 2017; 595:4207-4225. [PMID: 28295363 DOI: 10.1113/jp273883] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 03/10/2017] [Indexed: 12/25/2022] Open
Abstract
KEY POINTS Angiotensin II (AngII) is crucial in cardiovascular regulation in perinatal mammalians. Here we show that AngII increases twitch Ca2+ transients of mouse immature but not mature cardiomyocytes by robustly activating CaV 1.2 L-type Ca2+ channels through a novel signalling pathway involving angiotensin type 1 (AT1 ) receptors, β-arrestin2 and casein kinase 2. A β-arrestin-biased AT1 receptor agonist, TRV027, was as effective as AngII in activating L-type Ca2+ channels. Our results help understand the molecular mechanism by which AngII regulates the perinatal circulation and also suggest that β-arrestin-biased AT1 receptor agonists may be valuable therapeutics for paediatric heart failure. ABSTRACT Angiotensin II (AngII), the main effector peptide of the renin-angiotensin system, plays important roles in cardiovascular regulation in the perinatal period. Despite the well-known stimulatory effect of AngII on vascular contraction, little is known about regulation of contraction of the immature heart by AngII. Here we found that AngII significantly increased the peak amplitude of twitch Ca2+ transients by robustly activating L-type CaV 1.2 Ca2+ (CaV 1.2) channels in mouse immature but not mature cardiomyocytes. This response to AngII was mediated by AT1 receptors and β-arrestin2. A β-arrestin-biased AT1 receptor agonist was as effective as AngII in activating CaV 1.2 channels. Src-family tyrosine kinases (SFKs) and casein kinase 2α'β (CK2α'β) were sequentially activated when AngII activated CaV 1.2 channels. A cyclin-dependent kinase inhibitor, p27Kip1 (p27), inhibited CK2α'β, and AngII removed this inhibitory effect through phosphorylating tyrosine 88 of p27 via SFKs in cardiomyocytes. In a human embryonic kidney cell line, tsA201 cells, overexpression of CK2α'β but not c-Src directly activated recombinant CaV 1.2 channels composed of C-terminally truncated α1C , the distal C-terminus of α1C , β2C and α2 δ1 subunits, by phosphorylating threonine 1704 located at the interface between the proximal and the distal C-terminus of CaV 1.2α1C subunits. Co-immunoprecipitation revealed that CaV 1.2 channels, CK2α'β and p27 formed a macromolecular complex. Therefore, stimulation of AT1 receptors by AngII activates CaV 1.2 channels through β-arrestin2 and CK2α'β, thereby probably exerting a positive inotropic effect in the immature heart. Our results also indicated that β-arrestin-biased AT1 receptor agonists may be used as valuable therapeutics for paediatric heart failure in the future.
Collapse
Affiliation(s)
- Toshihide Kashihara
- Department of Molecular Pharmacology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Tsutomu Nakada
- Department of Molecular Pharmacology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Katsuhiko Kojima
- Department of Microbiology and Immunology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Toshikazu Takeshita
- Department of Microbiology and Immunology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Mitsuhiko Yamada
- Department of Molecular Pharmacology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| |
Collapse
|
27
|
Tykocki NR, Boerman EM, Jackson WF. Smooth Muscle Ion Channels and Regulation of Vascular Tone in Resistance Arteries and Arterioles. Compr Physiol 2017; 7:485-581. [PMID: 28333380 DOI: 10.1002/cphy.c160011] [Citation(s) in RCA: 233] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Vascular tone of resistance arteries and arterioles determines peripheral vascular resistance, contributing to the regulation of blood pressure and blood flow to, and within the body's tissues and organs. Ion channels in the plasma membrane and endoplasmic reticulum of vascular smooth muscle cells (SMCs) in these blood vessels importantly contribute to the regulation of intracellular Ca2+ concentration, the primary determinant of SMC contractile activity and vascular tone. Ion channels provide the main source of activator Ca2+ that determines vascular tone, and strongly contribute to setting and regulating membrane potential, which, in turn, regulates the open-state-probability of voltage gated Ca2+ channels (VGCCs), the primary source of Ca2+ in resistance artery and arteriolar SMCs. Ion channel function is also modulated by vasoconstrictors and vasodilators, contributing to all aspects of the regulation of vascular tone. This review will focus on the physiology of VGCCs, voltage-gated K+ (KV) channels, large-conductance Ca2+-activated K+ (BKCa) channels, strong-inward-rectifier K+ (KIR) channels, ATP-sensitive K+ (KATP) channels, ryanodine receptors (RyRs), inositol 1,4,5-trisphosphate receptors (IP3Rs), and a variety of transient receptor potential (TRP) channels that contribute to pressure-induced myogenic tone in resistance arteries and arterioles, the modulation of the function of these ion channels by vasoconstrictors and vasodilators, their role in the functional regulation of tissue blood flow and their dysfunction in diseases such as hypertension, obesity, and diabetes. © 2017 American Physiological Society. Compr Physiol 7:485-581, 2017.
Collapse
Affiliation(s)
- Nathan R Tykocki
- Department of Pharmacology, University of Vermont, Burlington, Vermont, USA
| | - Erika M Boerman
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
| | - William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
28
|
Olde Loohuis NFM, Nadif Kasri N, Glennon JC, van Bokhoven H, Hébert SS, Kaplan BB, Martens GJM, Aschrafi A. The schizophrenia risk gene MIR137 acts as a hippocampal gene network node orchestrating the expression of genes relevant to nervous system development and function. Prog Neuropsychopharmacol Biol Psychiatry 2017; 73:109-118. [PMID: 26925706 PMCID: PMC5002268 DOI: 10.1016/j.pnpbp.2016.02.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/08/2016] [Accepted: 02/21/2016] [Indexed: 02/06/2023]
Abstract
MicroRNAs (miRs) are small regulatory molecules, which orchestrate neuronal development and plasticity through modulation of complex gene networks. MicroRNA-137 (miR-137) is a brain-enriched RNA with a critical role in regulating brain development and in mediating synaptic plasticity. Importantly, mutations in this miR are associated with the pathoetiology of schizophrenia (SZ), and there is a widespread assumption that disruptions in miR-137 expression lead to aberrant expression of gene regulatory networks associated with SZ. To systematically identify the mRNA targets for this miR, we performed miR-137 gain- and loss-of-function experiments in primary rat hippocampal neurons and profiled differentially expressed mRNAs through next-generation sequencing. We identified 500 genes that were bidirectionally activated or repressed in their expression by the modulation of miR-137 levels. Gene ontology analysis using two independent software resources suggested functions for these miR-137-regulated genes in neurodevelopmental processes, neuronal maturation processes and cell maintenance, all of which known to be critical for proper brain circuitry formation. Since many of the putative miR-137 targets identified here also have been previously shown to be associated with SZ, we propose that this miR acts as a critical gene network hub contributing to the pathophysiology of this neurodevelopmental disorder.
Collapse
Affiliation(s)
- Nikkie F M Olde Loohuis
- Department of Cognitive Neuroscience, Radboudumc, 6500 HB Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ Nijmegen, The Netherlands
| | - Nael Nadif Kasri
- Department of Cognitive Neuroscience, Radboudumc, 6500 HB Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ Nijmegen, The Netherlands; Department of Human Genetics, Radboudumc, 6500 HB Nijmegen, The Netherlands
| | - Jeffrey C Glennon
- Department of Cognitive Neuroscience, Radboudumc, 6500 HB Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ Nijmegen, The Netherlands
| | - Hans van Bokhoven
- Department of Cognitive Neuroscience, Radboudumc, 6500 HB Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ Nijmegen, The Netherlands; Department of Human Genetics, Radboudumc, 6500 HB Nijmegen, The Netherlands
| | - Sébastien S Hébert
- Axe Neurosciences, Centre de recherche du CHU de Québec, CHUL, Québec, QC G1V4G2, Canada; Département de psychiatrie et neurosciences, Université Laval, Québec, QC G1V 0A6, Canada
| | - Barry B Kaplan
- Laboratory of Molecular Biology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gerard J M Martens
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ Nijmegen, The Netherlands; Department of Molecular Animal Physiology, Radboud University Nijmegen, 6525 HP Nijmegen, The Netherlands
| | - Armaz Aschrafi
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ Nijmegen, The Netherlands; Laboratory of Molecular Biology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
29
|
Jacquemet G, Baghirov H, Georgiadou M, Sihto H, Peuhu E, Cettour-Janet P, He T, Perälä M, Kronqvist P, Joensuu H, Ivaska J. L-type calcium channels regulate filopodia stability and cancer cell invasion downstream of integrin signalling. Nat Commun 2016; 7:13297. [PMID: 27910855 PMCID: PMC5146291 DOI: 10.1038/ncomms13297] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 09/19/2016] [Indexed: 01/03/2023] Open
Abstract
Mounting in vitro, in vivo and clinical evidence suggest an important role for filopodia in driving cancer cell invasion. Using a high-throughput microscopic-based drug screen, we identify FDA-approved calcium channel blockers (CCBs) as potent inhibitors of filopodia formation in cancer cells. Unexpectedly, we discover that L-type calcium channels are functional and frequently expressed in cancer cells suggesting a previously unappreciated role for these channels during tumorigenesis. We further demonstrate that, at filopodia, L-type calcium channels are activated by integrin inside-out signalling, integrin activation and Src. Moreover, L-type calcium channels promote filopodia stability and maturation into talin-rich adhesions through the spatially restricted regulation of calcium entry and subsequent activation of the protease calpain-1. Altogether we uncover a novel and clinically relevant signalling pathway that regulates filopodia formation in cancer cells and propose that cycles of filopodia stabilization, followed by maturation into focal adhesions, directs cancer cell migration and invasion. Filopodia have a prominent role in driving cancer cell invasion. Here, the authors show that L-type calcium channels are a druggable target regulating filopodia stability and maturation into focal adhesions in metastatic breast cancer cells.
Collapse
Affiliation(s)
- Guillaume Jacquemet
- Turku Centre for Biotechnology, University of Turku, FIN-20520 Turku, Finland
| | - Habib Baghirov
- Turku Centre for Biotechnology, University of Turku, FIN-20520 Turku, Finland
| | - Maria Georgiadou
- Turku Centre for Biotechnology, University of Turku, FIN-20520 Turku, Finland
| | - Harri Sihto
- Laboratory of Molecular Oncology, Translational Cancer Biology program, University of Helsinki, FIN-00290 Helsinki, Finland
| | - Emilia Peuhu
- Turku Centre for Biotechnology, University of Turku, FIN-20520 Turku, Finland
| | | | - Tao He
- VTT Medical Biotechnology, Technical Research Centre of Finland, FIN-20520 Turku, Finland
| | - Merja Perälä
- VTT Medical Biotechnology, Technical Research Centre of Finland, FIN-20520 Turku, Finland
| | - Pauliina Kronqvist
- Department of Pathology, University of Turku and Turku University Hospital, FIN-20520 Turku, Finland
| | - Heikki Joensuu
- Laboratory of Molecular Oncology, Translational Cancer Biology program, University of Helsinki, FIN-00290 Helsinki, Finland.,Department of Oncology, Helsinki University Hospital, FIN-00290 Helsinki, Finland
| | - Johanna Ivaska
- Turku Centre for Biotechnology, University of Turku, FIN-20520 Turku, Finland.,Department of Biochemistry, University of Turku, FIN-20520 Turku, Finland
| |
Collapse
|
30
|
Ferrari LF, Araldi D, Bogen O, Levine JD. Extracellular matrix hyaluronan signals via its CD44 receptor in the increased responsiveness to mechanical stimulation. Neuroscience 2016; 324:390-8. [PMID: 26996509 DOI: 10.1016/j.neuroscience.2016.03.032] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 02/18/2016] [Accepted: 03/14/2016] [Indexed: 01/08/2023]
Abstract
We propose that the extracellular matrix (ECM) signals CD44, a hyaluronan receptor, to increase the responsiveness to mechanical stimulation in the rat hind paw. We report that intradermal injection of hyaluronidase induces mechanical hyperalgesia, that is inhibited by co-administration of a CD44 receptor antagonist, A5G27. The intradermal injection of low (LMWH) but not high (HMWH) molecular weight hyaluronan also induces mechanical hyperalgesia, an effect that was attenuated by pretreatment with HMWH or A5G27. Pretreatment with HMWH also attenuated the hyperalgesia induced by hyaluronidase. Similarly, intradermal injection of A6, a CD44 receptor agonist, produced hyperalgesia that was inhibited by HMWH and A5G27. Inhibitors of protein kinase A (PKA) and Src, but not protein kinase C (PKC), significantly attenuated the hyperalgesia induced by both A6 and LMWH. Finally, to determine if CD44 receptor signaling is involved in a preclinical model of inflammatory pain, we evaluated the effect of A5G27 and HMWH on the mechanical hyperalgesia associated with the inflammation induced by carrageenan. Both A5G27 and HMWH attenuated carrageenan-induced mechanical hyperalgesia. Thus, while LMWH acts at its cognate receptor, CD44, to induce mechanical hyperalgesia, HMWH acts at the same receptor as an antagonist. That the local administration of HMWH or A5G27 inhibits carrageenan-induced hyperalgesia supports the suggestion that carrageenan produces changes in the ECM that contributes to inflammatory pain. These studies define a clinically relevant role for signaling by the hyaluronan receptor, CD44, in increased responsiveness to mechanical stimulation.
Collapse
Affiliation(s)
- L F Ferrari
- Departments of Medicine and Oral Surgery, and Division of Neuroscience, University of California at San Francisco, 521 Parnassus Avenue, San Francisco, CA 94143, USA.
| | - D Araldi
- Departments of Medicine and Oral Surgery, and Division of Neuroscience, University of California at San Francisco, 521 Parnassus Avenue, San Francisco, CA 94143, USA.
| | - O Bogen
- Departments of Medicine and Oral Surgery, and Division of Neuroscience, University of California at San Francisco, 521 Parnassus Avenue, San Francisco, CA 94143, USA.
| | - J D Levine
- Departments of Medicine and Oral Surgery, and Division of Neuroscience, University of California at San Francisco, 521 Parnassus Avenue, San Francisco, CA 94143, USA.
| |
Collapse
|
31
|
Modulation of CaV1.2 calcium channel by neuropeptide W regulates vascular myogenic tone via G protein-coupled receptor 7. J Hypertens 2015; 33:2431-42. [DOI: 10.1097/hjh.0000000000000723] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
32
|
Shaifta Y, Irechukwu N, Prieto-Lloret J, MacKay CE, Marchon KA, Ward JPT, Knock GA. Divergent modulation of Rho-kinase and Ca(2+) influx pathways by Src family kinases and focal adhesion kinase in airway smooth muscle. Br J Pharmacol 2015; 172:5265-80. [PMID: 26294392 PMCID: PMC4864488 DOI: 10.1111/bph.13313] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 08/02/2015] [Accepted: 08/19/2015] [Indexed: 02/06/2023] Open
Abstract
Background and Purpose The importance of tyrosine kinases in airway smooth muscle (ASM) contraction is not fully understood. The aim of this study was to investigate the role of Src‐family kinases (SrcFK) and focal adhesion kinase (FAK) in GPCR‐mediated ASM contraction and associated signalling events. Experimental Approach Contraction was recorded in intact or α‐toxin permeabilized rat bronchioles. Phosphorylation of SrcFK, FAK, myosin light‐chain‐20 (MLC20) and myosin phosphatase targeting subunit‐1 (MYPT‐1) was evaluated in cultured human ASM cells (hASMC). [Ca2+]i was evaluated in Fura‐2 loaded hASMC. Responses to carbachol (CCh) and bradykinin (BK) and the contribution of SrcFK and FAK to these responses were determined. Key Results Contractile responses in intact bronchioles were inhibited by antagonists of SrcFK, FAK and Rho‐kinase, while after α‐toxin permeabilization, they were sensitive to inhibition of SrcFK and Rho‐kinase, but not FAK. CCh and BK increased phosphorylation of MYPT‐1 and MLC20 and auto‐phosphorylation of SrcFK and FAK. MYPT‐1 phosphorylation was sensitive to inhibition of Rho‐kinase and SrcFK, but not FAK. Contraction induced by SR Ca2+ depletion and equivalent [Ca2+]i responses in hASMC were sensitive to inhibition of both SrcFK and FAK, while depolarization‐induced contraction was sensitive to FAK inhibition only. SrcFK auto‐phosphorylation was partially FAK‐dependent, while FAK auto‐phosphorylation was SrcFK‐independent. Conclusions and Implications SrcFK mediates Ca2+‐sensitization in ASM, while SrcFK and FAK together and individually influence multiple Ca2+ influx pathways. Tyrosine phosphorylation is therefore a key upstream signalling event in ASM contraction and may be a viable target for modulating ASM tone in respiratory disease.
Collapse
Affiliation(s)
- Yasin Shaifta
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Nneka Irechukwu
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Jesus Prieto-Lloret
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Charles E MacKay
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Keisha A Marchon
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Jeremy P T Ward
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Greg A Knock
- Division of Asthma, Allergy and Lung Biology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| |
Collapse
|
33
|
Mufti RE, Zechariah A, Sancho M, Mazumdar N, Brett SE, Welsh DG. Implications of αvβ3 Integrin Signaling in the Regulation of Ca2+ Waves and Myogenic Tone in Cerebral Arteries. Arterioscler Thromb Vasc Biol 2015; 35:2571-8. [PMID: 26494230 DOI: 10.1161/atvbaha.115.305619] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 10/09/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The myogenic response is central to blood flow regulation in the brain. Its induction is tied to elevated cytosolic [Ca(2+)], a response primarily driven by voltage-gated Ca(2+) channels and secondarily by Ca(2+) wave production. Although the signaling events leading to the former are well studied, those driving Ca(2+) waves remain uncertain. APPROACH AND RESULTS We postulated that αvβ3 integrin signaling is integral to the generation of pressure-induced Ca(2+) waves and cerebral arterial tone. This hypothesis was tested in rat cerebral arteries using the synergistic strengths of pressure myography, rapid Ca(2+) imaging, and Western blot analysis. GRGDSP, a peptide that preferentially blocks αvβ3 integrin, attenuated myogenic tone, indicating the modest role for sarcoplasmic reticulum Ca(2+) release in myogenic tone generation. The RGD peptide was subsequently shown to impair Ca(2+) wave generation and myosin light chain 20 (MLC20) phosphorylation, the latter of which was attributed to the modulation of MLC kinase and MLC phosphatase via MYPT1-T855 phosphorylation. Subsequent experiments revealed that elevated pressure enhanced phospholipase Cγ1 phosphorylation in an RGD-dependent manner and that phospholipase C inhibition attenuated Ca(2+) wave generation. Direct inhibition of inositol 1, 4, 5-triphosphate receptors also impaired Ca(2+) wave generation, myogenic tone, and MLC20 phosphorylation, partly through the T-855 phosphorylation site of MYPT1. CONCLUSIONS Our investigation reveals a hitherto unknown role for αvβ3 integrin as a cerebral arterial pressure sensor. The membrane receptor facilitates Ca(2+) wave generation through a signaling cascade, involving phospholipase Cγ1, inositol 1,3,4 triphosphate production, and inositol 1, 4, 5-triphosphate receptor activation. These discrete asynchronous Ca(2+) events facilitate MLC20 phosphorylation and, in part, myogenic tone by influencing both MLC kinase and MLC phosphatase activity.
Collapse
Affiliation(s)
- Rania E Mufti
- From the Hotchkiss Brain Institute (R.E.M., A.Z., M.S., N.M., S.E.B., D.G.W.), Libin Cardiovascular Institute (R.E.M., A.Z., M.S., N.M., S.E.B., D.G.W.), and Department of Physiology and Pharmacology (R.E.M., A.Z., M.S., N.M., S.E.B., D.G.W.), University of Calgary, Alberta, Canada; and Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada (A.Z., M.S., N.M., S.E.B., D.G.W.)
| | - Anil Zechariah
- From the Hotchkiss Brain Institute (R.E.M., A.Z., M.S., N.M., S.E.B., D.G.W.), Libin Cardiovascular Institute (R.E.M., A.Z., M.S., N.M., S.E.B., D.G.W.), and Department of Physiology and Pharmacology (R.E.M., A.Z., M.S., N.M., S.E.B., D.G.W.), University of Calgary, Alberta, Canada; and Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada (A.Z., M.S., N.M., S.E.B., D.G.W.)
| | - Maria Sancho
- From the Hotchkiss Brain Institute (R.E.M., A.Z., M.S., N.M., S.E.B., D.G.W.), Libin Cardiovascular Institute (R.E.M., A.Z., M.S., N.M., S.E.B., D.G.W.), and Department of Physiology and Pharmacology (R.E.M., A.Z., M.S., N.M., S.E.B., D.G.W.), University of Calgary, Alberta, Canada; and Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada (A.Z., M.S., N.M., S.E.B., D.G.W.)
| | - Neil Mazumdar
- From the Hotchkiss Brain Institute (R.E.M., A.Z., M.S., N.M., S.E.B., D.G.W.), Libin Cardiovascular Institute (R.E.M., A.Z., M.S., N.M., S.E.B., D.G.W.), and Department of Physiology and Pharmacology (R.E.M., A.Z., M.S., N.M., S.E.B., D.G.W.), University of Calgary, Alberta, Canada; and Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada (A.Z., M.S., N.M., S.E.B., D.G.W.)
| | - Suzanne E Brett
- From the Hotchkiss Brain Institute (R.E.M., A.Z., M.S., N.M., S.E.B., D.G.W.), Libin Cardiovascular Institute (R.E.M., A.Z., M.S., N.M., S.E.B., D.G.W.), and Department of Physiology and Pharmacology (R.E.M., A.Z., M.S., N.M., S.E.B., D.G.W.), University of Calgary, Alberta, Canada; and Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada (A.Z., M.S., N.M., S.E.B., D.G.W.)
| | - Donald G Welsh
- From the Hotchkiss Brain Institute (R.E.M., A.Z., M.S., N.M., S.E.B., D.G.W.), Libin Cardiovascular Institute (R.E.M., A.Z., M.S., N.M., S.E.B., D.G.W.), and Department of Physiology and Pharmacology (R.E.M., A.Z., M.S., N.M., S.E.B., D.G.W.), University of Calgary, Alberta, Canada; and Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada (A.Z., M.S., N.M., S.E.B., D.G.W.).
| |
Collapse
|
34
|
Dzwonek J, Wilczynski GM. CD44: molecular interactions, signaling and functions in the nervous system. Front Cell Neurosci 2015; 9:175. [PMID: 25999819 PMCID: PMC4423434 DOI: 10.3389/fncel.2015.00175] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 04/20/2015] [Indexed: 01/09/2023] Open
Abstract
CD44 is the major surface hyaluronan (HA) receptor implicated in intercellular and cell-matrix adhesion, cell migration and signaling. It is a transmembrane, highly glycosylated protein with several isoforms resulting from alternative gene splicing. The CD44 molecule consists of several domains serving different functions: the N-terminal extracellular domain, the stem region, the transmembrane domain and the C-terminal tail. In the nervous system, CD44 expression occurs in both glial and neuronal cells. The role of CD44 in the physiology and pathology of the nervous system is not entirely understood, however, there exists evidence suggesting it might be involved in the axon guidance, cytoplasmic Ca2+ clearance, dendritic arborization, synaptic transmission, epileptogenesis, oligodendrocyte and astrocyte differentiation, post-traumatic brain repair and brain tumour development.
Collapse
Affiliation(s)
- Joanna Dzwonek
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology Warsaw, Poland
| | - Grzegorz M Wilczynski
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology Warsaw, Poland
| |
Collapse
|
35
|
Zhang Y, Ying J, Jiang D, Chang Z, Li H, Zhang G, Gong S, Jiang X, Tao J. Urotensin-II receptor stimulation of cardiac L-type Ca2+ channels requires the βγ subunits of Gi/o-protein and phosphatidylinositol 3-kinase-dependent protein kinase C β1 isoform. J Biol Chem 2015; 290:8644-55. [PMID: 25678708 DOI: 10.1074/jbc.m114.615021] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Recent studies have demonstrated that urotensin-II (U-II) plays important roles in cardiovascular actions including cardiac positive inotropic effects and increasing cardiac output. However, the mechanisms underlying these effects of U-II in cardiomyocytes still remain unknown. We show by electrophysiological studies that U-II dose-dependently potentiates L-type Ca(2+) currents (ICa,L) in adult rat ventricular myocytes. This effect was U-II receptor (U-IIR)-dependent and was associated with a depolarizing shift in the voltage dependence of inactivation. Intracellular application of guanosine-5'-O-(2-thiodiphosphate) and pertussis toxin pretreatment both abolished the stimulatory effects of U-II. Dialysis of cells with the QEHA peptide, but not scrambled peptide SKEE, blocked the U-II-induced response. The phosphatidylinositol 3-kinase (PI3K) inhibitor wortmannin as well as the class I PI3K antagonist CH132799 blocked the U-II-induced ICa,L response. Protein kinase C antagonists calphostin C and chelerythrine chloride as well as dialysis of cells with 1,2bis(2aminophenoxy)ethaneN,N,N',N'-tetraacetic acid abolished the U-II-induced responses, whereas PKCα inhibition or PKA blockade had no effect. Exposure of ventricular myocytes to U-II markedly increased membrane PKCβ1 expression, whereas inhibition of PKCβ1 pharmacologically or by shRNA targeting abolished the U-II-induced ICa,L response. Functionally, we observed a significant increase in the amplitude of sarcomere shortening induced by U-II; blockade of U-IIR as well as PKCβ inhibition abolished this effect, whereas Bay K8644 mimicked the U-II response. Taken together, our results indicate that U-II potentiates ICa,L through the βγ subunits of Gi/o-protein and downstream activation of the class I PI3K-dependent PKCβ1 isoform. This occurred via the activation of U-IIR and contributes to the positive inotropic effect on cardiomyocytes.
Collapse
Affiliation(s)
- Yuan Zhang
- From the Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou 215123, China, Department of Geriatrics and Institute of Neuroscience, the Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Jiaoqian Ying
- From the Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou 215123, China, Department of Emergency Medicine, China-Japan Friendship Hospital, Beijing 100029, China
| | - Dongsheng Jiang
- From the Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou 215123, China, Department of Dermatology and Allergic Diseases, University of Ulm, Ulm 89081, Germany, and
| | - Zhigang Chang
- From the Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou 215123, China
| | - Hua Li
- From the Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou 215123, China, National Shanghai Center for New Drug Safety Evaluation and Research, Shanghai 201203, China
| | - Guoqiang Zhang
- Department of Emergency Medicine, China-Japan Friendship Hospital, Beijing 100029, China
| | - Shan Gong
- From the Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou 215123, China
| | - Xinghong Jiang
- From the Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou 215123, China
| | - Jin Tao
- From the Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou 215123, China,
| |
Collapse
|
36
|
Chen W, Ke JB, Wu HJ, Miao Y, Li F, Yang XL, Wang Z. Somatostatin receptor-mediated suppression of gabaergic synaptic transmission in cultured rat retinal amacrine cells. Neuroscience 2014; 273:118-27. [PMID: 24846611 DOI: 10.1016/j.neuroscience.2014.05.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 05/05/2014] [Accepted: 05/06/2014] [Indexed: 01/03/2023]
Abstract
Somatostatin (SRIF) modulates neurotransmitter release by activating the specific receptors (sst1-sst5). Our previous study showed that sst5 receptors are expressed in rat retinal GABAergic amacrine cells. Here, we investigated modulation of GABA release by SRIF in cultured amacrine cells, using patch-clamp techniques. The frequency of spontaneous inhibitory postsynaptic currents (sIPSCs) in the amacrine cells was significantly reduced by SRIF, which was partially reversed by BIM 23056, an sst5 receptor antagonist, and was further rescued by addition of CYN-154806, an sst2 receptor antagonist. Both nimodipine, an L-type Ca2+ channel blocker, and ω-conotoxin GVIA, an N-type Ca2+ channel blocker, suppressed the sIPSC frequency, and in the presence of nimodipine and ω-conotoxin GVIA, SRIF failed to further suppress the sIPSC frequency. Extracellular application of forskolin, an activator of adenylate cyclase, increased the sIPSC frequency, while the membrane permeable protein kinase A (PKA) inhibitor Rp-cAMP reduced it, and in the presence of Rp-cAMP, SRIF did not change sIPSCs. However, SRIF persisted to suppress the sIPSCs in the presence of KT5823, a protein kinase G (PKG) inhibitor. Moreover, pre-incubation with Bis IV, a protein kinase C (PKC) inhibitor, or pre-application of xestospongin C, an inositol 1,4,5-trisphosphate receptor (IP3R) inhibitor, SRIF still suppressed the sIPSC frequency. All these results suggest that SRIF suppresses GABA release from the amacrine cells by inhibiting presynaptic Ca2+ channels, in part through activating sst5/sst2 receptors, a process that is mediated by the intracellular cAMP-PKA signaling pathway.
Collapse
Affiliation(s)
- W Chen
- Institutes of Brain Science, Institute of Neurobiology and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - J B Ke
- Institutes of Brain Science, Institute of Neurobiology and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - H J Wu
- Institutes of Brain Science, Institute of Neurobiology and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - Y Miao
- Institutes of Brain Science, Institute of Neurobiology and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - F Li
- Institutes of Brain Science, Institute of Neurobiology and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - X L Yang
- Institutes of Brain Science, Institute of Neurobiology and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - Z Wang
- Institutes of Brain Science, Institute of Neurobiology and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China.
| |
Collapse
|
37
|
CaV1.3 L-type channels, maxiK Ca2+-dependent K+ channels and bestrophin-1 regulate rhythmic photoreceptor outer segment phagocytosis by retinal pigment epithelial cells. Cell Signal 2014; 26:968-78. [DOI: 10.1016/j.cellsig.2013.12.021] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 12/31/2013] [Indexed: 11/20/2022]
|
38
|
Xia M, Zhu Y. Fibronectin enhances spinal cord astrocyte proliferation by elevating P2Y1 receptor expression. J Neurosci Res 2014; 92:1078-90. [PMID: 24687862 DOI: 10.1002/jnr.23384] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Revised: 02/16/2014] [Accepted: 02/19/2014] [Indexed: 12/15/2022]
Affiliation(s)
- Maosheng Xia
- Department of Orthopaedics; The First Hospital of China Medical University; Shengyang People's Republic of China
| | - Yue Zhu
- Department of Orthopaedics; The First Hospital of China Medical University; Shengyang People's Republic of China
| |
Collapse
|
39
|
Abstract
Integrins are heterodimeric, transmembrane receptors that are expressed in all cells, including those in the heart. They participate in multiple critical cellular processes including adhesion, extracellular matrix organization, signaling, survival, and proliferation. Particularly relevant for a contracting muscle cell, integrins are mechanotransducers, translating mechanical to biochemical information. Although it is likely that cardiovascular clinicians and scientists have the highest recognition of integrins in the cardiovascular system from drugs used to inhibit platelet aggregation, the focus of this article will be on the role of integrins specifically in the cardiac myocyte. After a general introduction to integrin biology, the article will discuss important work on integrin signaling, mechanotransduction, and lessons learned about integrin function from a range of model organisms. Then we will detail work on integrin-related proteins in the myocyte, how integrins may interact with ion channels and mediate viral uptake into cells, and also play a role in stem cell biology. Finally, we will discuss directions for future study.
Collapse
Affiliation(s)
- Sharon Israeli-Rosenberg
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA, USA, and Veterans Administration San Diego Healthcare System, San Diego, CA, USA
| | - Ana Maria Manso
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA, USA, and Veterans Administration San Diego Healthcare System, San Diego, CA, USA
| | - Hideshi Okada
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA, USA, and Veterans Administration San Diego Healthcare System, San Diego, CA, USA
| | - Robert S Ross
- Department of Medicine, Cardiology, UCSD School of Medicine, La Jolla, CA, USA, and Veterans Administration San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
40
|
Luo J, McGinnis LK, Carlton C, Beggs HE, Kinsey WH. PTK2b function during fertilization of the mouse oocyte. Biochem Biophys Res Commun 2014; 450:1212-7. [PMID: 24667605 DOI: 10.1016/j.bbrc.2014.03.083] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 03/11/2014] [Indexed: 12/12/2022]
Abstract
Fertilization triggers rapid changes in intracellular free calcium that serve to activate multiple signaling events critical to the initiation of successful development. Among the pathways downstream of the fertilization-induced calcium transient is the calcium-calmodulin dependent protein tyrosine kinase PTK2b or PYK2 kinase. PTK2b plays an important role in fertilization of the zebrafish oocyte and the objective of the present study was to establish whether PTK2b also functions in mammalian fertilization. PTK2b was activated during the first few hours after fertilization of the mouse oocyte during the period when anaphase resumption was underway and prior to the pronuclear stage. Suppression of PTK2b kinase activity in oocytes blocked sperm incorporation and egg activation although sperm-oocyte binding was not affected. Oocytes that failed to incorporate sperm after inhibitor treatment showed no evidence of a calcium transient and no evidence of anaphase resumption suggesting that egg activation did not occur. The results indicate that PTK2b functions during the sperm-egg fusion process or during the physical incorporation of sperm into the egg cytoplasm and is therefore critical for successful development.
Collapse
Affiliation(s)
- Jinping Luo
- Department of Anatomy & Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Lynda K McGinnis
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Carol Carlton
- Department of Anatomy & Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Hilary E Beggs
- Department of Ophthalmology, University of California, San Francisco, CA, United States
| | - William H Kinsey
- Department of Anatomy & Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, United States.
| |
Collapse
|
41
|
Sheng L, Leshchyns'ka I, Sytnyk V. Cell adhesion and intracellular calcium signaling in neurons. Cell Commun Signal 2013; 11:94. [PMID: 24330678 PMCID: PMC3878801 DOI: 10.1186/1478-811x-11-94] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 12/05/2013] [Indexed: 01/10/2023] Open
Abstract
Cell adhesion molecules (CAMs) play indispensable roles in the developing and mature brain by regulating neuronal migration and differentiation, neurite outgrowth, axonal fasciculation, synapse formation and synaptic plasticity. CAM-mediated changes in neuronal behavior depend on a number of intracellular signaling cascades including changes in various second messengers, among which CAM-dependent changes in intracellular Ca2+ levels play a prominent role. Ca2+ is an essential secondary intracellular signaling molecule that regulates fundamental cellular functions in various cell types, including neurons. We present a systematic review of the studies reporting changes in intracellular Ca2+ levels in response to activation of the immunoglobulin superfamily CAMs, cadherins and integrins in neurons. We also analyze current experimental evidence on the Ca2+ sources and channels involved in intracellular Ca2+ increases mediated by CAMs of these families, and systematically review the role of the voltage-dependent Ca2+ channels (VDCCs) in neurite outgrowth induced by activation of these CAMs. Molecular mechanisms linking CAMs to VDCCs and intracellular Ca2+ stores in neurons are discussed.
Collapse
Affiliation(s)
| | | | - Vladimir Sytnyk
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales 2052, Australia.
| |
Collapse
|
42
|
Stock C, Ludwig FT, Hanley PJ, Schwab A. Roles of ion transport in control of cell motility. Compr Physiol 2013; 3:59-119. [PMID: 23720281 DOI: 10.1002/cphy.c110056] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cell motility is an essential feature of life. It is essential for reproduction, propagation, embryonic development, and healing processes such as wound closure and a successful immune defense. If out of control, cell motility can become life-threatening as, for example, in metastasis or autoimmune diseases. Regardless of whether ciliary/flagellar or amoeboid movement, controlled motility always requires a concerted action of ion channels and transporters, cytoskeletal elements, and signaling cascades. Ion transport across the plasma membrane contributes to cell motility by affecting the membrane potential and voltage-sensitive ion channels, by inducing local volume changes with the help of aquaporins and by modulating cytosolic Ca(2+) and H(+) concentrations. Voltage-sensitive ion channels serve as voltage detectors in electric fields thus enabling galvanotaxis; local swelling facilitates the outgrowth of protrusions at the leading edge while local shrinkage accompanies the retraction of the cell rear; the cytosolic Ca(2+) concentration exerts its main effect on cytoskeletal dynamics via motor proteins such as myosin or dynein; and both, the intracellular and the extracellular H(+) concentration modulate cell migration and adhesion by tuning the activity of enzymes and signaling molecules in the cytosol as well as the activation state of adhesion molecules at the cell surface. In addition to the actual process of ion transport, both, channels and transporters contribute to cell migration by being part of focal adhesion complexes and/or physically interacting with components of the cytoskeleton. The present article provides an overview of how the numerous ion-transport mechanisms contribute to the various modes of cell motility.
Collapse
Affiliation(s)
- Christian Stock
- Institute of Physiology II, University of Münster, Münster, Germany.
| | | | | | | |
Collapse
|
43
|
Shi Y, Yang G, Yu J, Yu L, Westenbroek R, Catterall WA, Juntti-Berggren L, Berggren PO, Yang SN. Apolipoprotein CIII hyperactivates β cell CaV1 channels through SR-BI/β1 integrin-dependent coactivation of PKA and Src. Cell Mol Life Sci 2013; 71:1289-303. [PMID: 23949443 DOI: 10.1007/s00018-013-1442-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 07/06/2013] [Accepted: 07/29/2013] [Indexed: 11/30/2022]
Abstract
Apolipoprotein CIII (ApoCIII) not only serves as an inhibitor of triglyceride hydrolysis but also participates in diabetes-related pathological events such as hyperactivation of voltage-gated Ca(2+) (CaV) channels in the pancreatic β cell. However, nothing is known about the molecular mechanisms whereby ApoCIII hyperactivates β cell CaV channels. We now demonstrate that ApoCIII increased CaV1 channel open probability and density. ApoCIII enhanced whole-cell Ca(2+) currents and the CaV1 channel blocker nimodipine completely abrogated this enhancement. The effect of ApoCIII was not influenced by individual inhibition of PKA, PKC, or Src. However, combined inhibition of PKA, PKC, and Src counteracted the effect of ApoCIII, similar results obtained by coinhibition of PKA and Src. Moreover, knockdown of β1 integrin or scavenger receptor class B type I (SR-BI) prevented ApoCIII from hyperactivating β cell CaV channels. These data reveal that ApoCIII hyperactivates β cell CaV1 channels through SR-BI/β1 integrin-dependent coactivation of PKA and Src.
Collapse
Affiliation(s)
- Yue Shi
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-171 76, Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Gulia J, Navedo MF, Gui P, Chao JT, Mercado JL, Santana LF, Davis MJ. Regulation of L-type calcium channel sparklet activity by c-Src and PKC-α. Am J Physiol Cell Physiol 2013; 305:C568-77. [PMID: 23804206 DOI: 10.1152/ajpcell.00381.2011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The activity of persistent Ca²⁺ sparklets, which are characterized by longer and more frequent channel open events than low-activity sparklets, contributes substantially to steady-state Ca²⁺ entry under physiological conditions. Here, we addressed two questions related to the regulation of Ca²⁺ sparklets by PKC-α and c-Src, both of which increase whole cell Cav1.2 current: 1) Does c-Src activation enhance persistent Ca²⁺ sparklet activity? 2) Does PKC-α activate c-Src to produce persistent Ca²⁺ sparklets? With the use of total internal reflection fluorescence microscopy, Ca²⁺ sparklets were recorded from voltage-clamped tsA-201 cells coexpressing wild-type (WT) or mutant Cav1.2c (the neuronal isoform of Cav1.2) constructs ± active or inactive PKC-α/c-Src. Cells expressing Cav1.2c exhibited both low-activity and persistent Ca²⁺ sparklets. Persistent Ca²⁺ sparklet activity was significantly reduced by acute application of the c-Src inhibitor PP2 or coexpression of kinase-dead c-Src. Cav1.2c constructs mutated at one of two COOH-terminal residues (Y²¹²²F and Y²¹³⁹F) were used to test the effect of blocking putative phosphorylation sites for c-Src. Expression of Y²¹²²F but not Y²¹³⁹F Cav1.2c abrogated the potentiating effect of c-Src on Ca²⁺ sparklet activity. We could not detect a significant change in persistent Ca²⁺ sparklet activity or density in cells coexpressing Cav1.2c + PKC-α, regardless of whether WT or Y²¹²²F Cav1.2c was used, or after PP2 application, suggesting that PKC-α does not act upstream of c-Src to produce persistent Ca²⁺ sparklets. However, our results indicate that persistent Ca²⁺ sparklet activity is promoted by the action of c-Src on residue Y²¹²² of the Cav1.2c COOH terminus.
Collapse
Affiliation(s)
- Jyoti Gulia
- Department of Biological Engineering University of Missouri, Columbia, Missouri, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Zhang CH, Lifshitz LM, Uy KF, Ikebe M, Fogarty KE, ZhuGe R. The cellular and molecular basis of bitter tastant-induced bronchodilation. PLoS Biol 2013; 11:e1001501. [PMID: 23472053 PMCID: PMC3589262 DOI: 10.1371/journal.pbio.1001501] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 01/24/2013] [Indexed: 01/13/2023] Open
Abstract
Bitter tastants can activate bitter taste receptors on constricted smooth muscle cells to inhibit L-type calcium channels and induce bronchodilation. Bronchodilators are a standard medicine for treating airway obstructive diseases, and β2 adrenergic receptor agonists have been the most commonly used bronchodilators since their discovery. Strikingly, activation of G-protein-coupled bitter taste receptors (TAS2Rs) in airway smooth muscle (ASM) causes a stronger bronchodilation in vitro and in vivo than β2 agonists, implying that new and better bronchodilators could be developed. A critical step towards realizing this potential is to understand the mechanisms underlying this bronchodilation, which remain ill-defined. An influential hypothesis argues that bitter tastants generate localized Ca2+ signals, as revealed in cultured ASM cells, to activate large-conductance Ca2+-activated K+ channels, which in turn hyperpolarize the membrane, leading to relaxation. Here we report that in mouse primary ASM cells bitter tastants neither evoke localized Ca2+ events nor alter spontaneous local Ca2+ transients. Interestingly, they increase global intracellular [Ca2+]i, although to a much lower level than bronchoconstrictors. We show that these Ca2+ changes in cells at rest are mediated via activation of the canonical bitter taste signaling cascade (i.e., TAS2R-gustducin-phospholipase Cβ [PLCβ]- inositol 1,4,5-triphosphate receptor [IP3R]), and are not sufficient to impact airway contractility. But activation of TAS2Rs fully reverses the increase in [Ca2+]i induced by bronchoconstrictors, and this lowering of the [Ca2+]i is necessary for bitter tastant-induced ASM cell relaxation. We further show that bitter tastants inhibit L-type voltage-dependent Ca2+ channels (VDCCs), resulting in reversal in [Ca2+]i, and this inhibition can be prevented by pertussis toxin and G-protein βγ subunit inhibitors, but not by the blockers of PLCβ and IP3R. Together, we suggest that TAS2R stimulation activates two opposing Ca2+ signaling pathways via Gβγ to increase [Ca2+]i at rest while blocking activated L-type VDCCs to induce bronchodilation of contracted ASM. We propose that the large decrease in [Ca2+]i caused by effective tastant bronchodilators provides an efficient cell-based screening method for identifying potent dilators from among the many thousands of available bitter tastants. Bitter taste receptors (TAS2Rs), a G-protein-coupled receptor family long thought to be solely expressed in taste buds on the tongue, have recently been detected in airways. Bitter substances can activate TAS2Rs in airway smooth muscle to cause greater bronchodilation than β2 adrenergic receptor agonists, the most commonly used bronchodilators. However, the mechanisms underlying this bronchodilation remain elusive. Here we show that, in resting primary airway smooth muscle cells, bitter tastants activate a TAS2R-dependent signaling pathway that results in an increase in intracellular calcium levels, albeit to a level much lower than that produced by bronchoconstrictors. In bronchoconstricted cells, however, bitter tastants reverse the bronchoconstrictor-induced increase in calcium levels, which leads to the relaxation of smooth muscle cells. We find that this reversal is due to inhibition of L-type calcium channels. Our results suggest that under normal conditions, bitter tastants can activate TAS2Rs to modestly increase calcium levels, but that when smooth muscle cells are constricted, they can block L-type calcium channels to induce bronchodilation. We postulate that this novel mechanism could operate in other extraoral cells expressing TAS2Rs.
Collapse
Affiliation(s)
- Cheng-Hai Zhang
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Lawrence M. Lifshitz
- Biomedical Imaging Group, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Karl F. Uy
- Department of Surgery, Division of Thoracic Surgery, University of Massachusetts Memorial Medical Center, Worcester, Massachusetts, United States of America
| | - Mitsuo Ikebe
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Kevin E. Fogarty
- Biomedical Imaging Group, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Ronghua ZhuGe
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Biomedical Imaging Group, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
46
|
Liu K, Gui B, Sun Y, Shi N, Gu Z, Zhang T, Sun X. Inhibition of L-type Ca(2+) channels by curcumin requires a novel protein kinase-theta isoform in rat hippocampal neurons. Cell Calcium 2012; 53:195-203. [PMID: 23261315 DOI: 10.1016/j.ceca.2012.11.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 10/18/2012] [Accepted: 11/19/2012] [Indexed: 11/26/2022]
Abstract
Curcumin, a major active compound of Curcuma longa, has been reported to have potent neuroprotective activities. However to date, the relevant mechanisms still remain unclear. In this study, we report that curcumin selectively inhibits L-type Ca(2+) channel currents in cultured rat hippocampal neurons. Whole-cell currents were recorded using 10mM barium as a charge carrier. Curcumin reversibly inhibited high-voltage-gated Ca(2+) channel (HVGCC) currents (IBa) in a concentration-dependent manner but had no apparent effects on the cells treated with nifedipine, a specific L-type Ca(2+) channel blocker. Curcumin did not markedly affect the activation of L-type Ca(2+) channels while significantly shifted the inactivation curve in the hyperpolarizing direction. Pretreatment of cells with the classical and novel PKC antagonists GF109203X and calphostin C completely abolished curcumin-induced IBa inhibition, whereas the classical PKC antagonist Gö6976 or inhibition of PKA activity elicited no such effects. Moreover, the curcumin-induced IBa response was abolished by intracellular application of the PKC-θ inhibitory peptide PKC-θ-IP or by siRNA knockdown of PKC-θ in cultured rat hippocampal neurons. In these neurons, novel isoforms of PKC including delta (PKC-δ), epsilon (PKC-ɛ) and theta (PKC-θ), but not eta (PKC-η), were endogenously expressed. Taken together, these results suggest that curcumin selectively inhibits IBavia a novel PKC-θ-dependent pathway, which could contribute to its neuroprotective effects in rat hippocampal neurons.
Collapse
Affiliation(s)
- Kangyong Liu
- Department of Neurology, Shanghai Pudong New Area Zhoupu Hospital, Shanghai 201138, PR China
| | | | | | | | | | | | | |
Collapse
|
47
|
Kinsey WH. Intersecting roles of protein tyrosine kinase and calcium signaling during fertilization. Cell Calcium 2012. [PMID: 23201334 DOI: 10.1016/j.ceca.2012.11.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The oocyte is a highly specialized cell that must respond to fertilization with a preprogrammed series of signal transduction events that establish a block to polyspermy, trigger resumption of the cell cycle and execution of a developmental program. The fertilization-induced calcium transient is a key signal that initiates the process of oocyte activation and studies over the last several years have examined the signaling pathways that act upstream and downstream of this calcium transient. Protein tyrosine kinase signaling was found to be an important component of the upstream pathways that stimulated calcium release at fertilization in oocytes from animals that fertilize externally, but a similar pathway has not been found in mammals which fertilize internally. The following review will examine the diversity of signaling in oocytes from marine invertebrates, amphibians, fish and mammals in an attempt to understand the basis for the observed differences. In addition to the pathways upstream of the fertilization-induced calcium transient, recent studies are beginning to unravel the role of protein tyrosine kinase signaling downstream of the calcium transient. The PYK2 kinase was found to respond to fertilization in the zebrafish system and seems to represent a novel component of the response of the oocyte to fertilization. The potential impact of impaired PTK signaling in oocyte quality will also be discussed.
Collapse
Affiliation(s)
- William H Kinsey
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, United States.
| |
Collapse
|
48
|
Vandael DHF, Mahapatra S, Calorio C, Marcantoni A, Carbone E. Cav1.3 and Cav1.2 channels of adrenal chromaffin cells: emerging views on cAMP/cGMP-mediated phosphorylation and role in pacemaking. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1828:1608-18. [PMID: 23159773 DOI: 10.1016/j.bbamem.2012.11.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 11/05/2012] [Accepted: 11/08/2012] [Indexed: 12/21/2022]
Abstract
Voltage-gated Ca²⁺ channels (VGCCs) are voltage sensors that convert membrane depolarizations into Ca²⁺ signals. In the chromaffin cells of the adrenal medulla, the Ca²⁺ signals driven by VGCCs regulate catecholamine secretion, vesicle retrievals, action potential shape and firing frequency. Among the VGCC-types expressed in these cells (N-, L-, P/Q-, R- and T-types), the two L-type isoforms, Ca(v)1.2 and Ca(v)1.3, control key activities due to their particular activation-inactivation gating and high-density of expression in rodents and humans. The two isoforms are also effectively modulated by G protein-coupled receptor pathways delimited in membrane micro-domains and by the cAMP/PKA and NO/cGMP/PKG phosphorylation pathways which induce prominent Ca²⁺ current changes if opposingly regulated. The two L-type isoforms shape the action potential and directly participate to vesicle exocytosis and endocytosis. The low-threshold of activation and slow rate of inactivation of Ca(v)1.3 confer to this channel the unique property of carrying sufficient inward current at subthreshold potentials able to activate BK and SK channels which set the resting potential, the action potential shape, the cell firing mode and the degree of spike frequency adaptation during spontaneous firing or sustained depolarizations. These properties help chromaffin cells to optimally adapt when switching from normal to stress-mimicking conditions. Here, we will review past and recent findings on cAMP- and cGMP-mediated modulations of Ca(v)1.2 and Ca(v)1.3 and the role that these channels play in the control of chromaffin cell firing. This article is part of a Special Issue entitled: Calcium channels.
Collapse
Affiliation(s)
- D H F Vandael
- Department of Drug Science, Laboratory of Cellular & Molecular Neuroscience, NIS Center, CNISM, University of Torino, Italy
| | | | | | | | | |
Collapse
|
49
|
Dabiri BE, Lee H, Parker KK. A potential role for integrin signaling in mechanoelectrical feedback. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2012; 110:196-203. [PMID: 22819851 DOI: 10.1016/j.pbiomolbio.2012.07.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 07/11/2012] [Indexed: 01/20/2023]
Abstract
Certain forms of heart disease involve gross morphological changes to the myocardium that alter its hemodynamic loading conditions. These changes can ultimately lead to the increased deposition of extracellular matrix (ECM) proteins, such as collagen and fibronectin, which together work to pathologically alter the myocardium's bulk tissue mechanics. In addition to changing the mechanical properties of the heart, this maladaptive remodeling gives rise to changes in myocardium electrical conductivity and synchrony since the tissue's mechanical properties are intimately tied to its electrical characteristics. This phenomenon, called mechanoelectrical coupling (MEC), can render individuals affected by heart disease arrhythmogenic and susceptible to Sudden Cardiac Death (SCD). The underlying mechanisms of MEC have been attributed to various processes, including the action of stretch activated channels and changes in troponin C-Ca(2+) binding affinity. However, changes in the heart post infarction or due to congenital myopathies are also accompanied by shifts in the expression of various molecular components of cardiomyocytes, including the mechanosensitive family of integrin proteins. As transmembrane proteins, integrins mechanically couple the ECM with the intracellular cytoskeleton and have been implicated in mediating ion homeostasis in various cell types, including neurons and smooth muscle. Given evidence of altered integrin expression in the setting of heart disease coupled with the associated increased risk for arrhythmia, we argue in this review that integrin signaling contributes to MEC. In light of the significant mortality associated with arrhythmia and SCD, close examination of all culpable mechanisms, including integrin-mediated MEC, is necessary.
Collapse
Affiliation(s)
- Borna E Dabiri
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard University, 29 Oxford St, Pierce Hall 321, Cambridge, MA 02138, USA
| | | | | |
Collapse
|
50
|
Wu X, Reddy DS. Integrins as receptor targets for neurological disorders. Pharmacol Ther 2011; 134:68-81. [PMID: 22233753 DOI: 10.1016/j.pharmthera.2011.12.008] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Accepted: 12/15/2011] [Indexed: 12/18/2022]
Abstract
This review focuses on the neurobiology of integrins, pathophysiological roles of integrins in neuroplasticity and nervous system disorders, and therapeutic implications of integrins as potential drug targets and possible delivery pathways. Neuroplasticity is a central phenomenon in many neurological conditions such as seizures, trauma, and traumatic brain injury. During the course of many brain diseases, in addition to intracellular compartment changes, alterations in non-cell compartments such as extracellular matrix (ECM) are recognized as an essential process in forming and reorganizing neural connections. Integrins are heterodimeric transmembrane receptors that mediate cell-ECM and cell-cell adhesion events. Although the mechanisms of neuroplasticity remain unclear, it has been suggested that integrins undergo plasticity including clustering through interactions with ECM proteins, modulating ion channels, intracellular Ca(2+) and protein kinase signaling, and reorganization of cytoskeletal filaments. As cell surface receptors, integrins are central to the pathophysiology of many brain diseases, such as epilepsy, and are potential targets for the development of new drugs for neurological disorders.
Collapse
Affiliation(s)
- Xin Wu
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Bryan, TX 77807, USA
| | | |
Collapse
|