1
|
Kumar D, Harris AL, Luo YL. Molecular permeation through large pore channels: computational approaches and insights. J Physiol 2024. [PMID: 39373834 DOI: 10.1113/jp285198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/06/2024] [Indexed: 10/08/2024] Open
Abstract
Computational methods such as molecular dynamics (MD) have illuminated how single-atom ions permeate membrane channels and how selectivity among them is achieved. Much less is understood about molecular permeation through eukaryotic channels that mediate the flux of small molecules (e.g. connexins, pannexins, LRRC8s, CALHMs). Here we describe computational methods that have been profitably employed to explore the movements of molecules through wide pores, revealing mechanistic insights, guiding experiments, and suggesting testable hypotheses. This review illustrates MD techniques such as voltage-driven flux, potential of mean force, and mean first-passage-time calculations, as applied to molecular permeation through wide pores. These techniques have enabled detailed and quantitative modeling of molecular interactions and movement of permeants at the atomic level. We highlight novel contributors to the transit of molecules through these wide pathways. In particular, the flexibility and anisotropic nature of permeant molecules, coupled with the dynamics of pore-lining residues, lead to bespoke permeation dynamics. As more eukaryotic large-pore channel structures and functional data become available, these insights and approaches will be important for understanding the physical principles underlying molecular permeation and as guides for experimental design.
Collapse
Affiliation(s)
- Deepak Kumar
- Department of Biotechnology and Pharmaceutical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Andrew L Harris
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Yun Lyna Luo
- Department of Biotechnology and Pharmaceutical Sciences, Western University of Health Sciences, Pomona, CA, USA
| |
Collapse
|
2
|
Skieresz-Szewczyk K, Jackowiak H. Pattern Distribution of Connexins in the Ortho- and Parakeratinized Epithelium of the Lingual Mucosa in Birds. Cells 2023; 12:1776. [PMID: 37443811 PMCID: PMC10341081 DOI: 10.3390/cells12131776] [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: 05/09/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Connexins are important proteins involved in cell-to-cell communication and cytodifferentiation during renewal and cornification of the multilayered epithelia. So far, there is a lack of reports on this subject in birds' structurally different ortho- and parakeratinized epithelium of the tongue. The study aims to describe the distribution and expression profiles of the α-connexins (Cx40 and 43) and β-connexins (Cx26, 30, and 31) in those epithelia in duck, goose, and domestic turkey. Research revealed the presence of the mentioned connexins and the occurrence of interspecies differences. Connexins form gap junctions in the cell membrane or are in the cytoplasm of keratinocytes. Differences in connexin expression were noted between the basal and intermediate layers, which may determine the proliferation of keratinocytes. Cx40, 43, and Cx30 in the gap junction of the keratinocytes of the intermediate layer are related to the synchronization of the cornification process. Because of the exfoliation of cornified plaques, a lack of connexins was observed in the cornified layer of orthokeratinized epithelium. However, in parakeratinized epithelium, connexins were present in the cell membrane of keratinocytes and thus maintained cellular integrity in gradually desquamating cells. The current studies will be useful in further comparative analyses of normal and pathological epithelia of the oral cavity in birds.
Collapse
Affiliation(s)
- Kinga Skieresz-Szewczyk
- Department of Histology and Embryology, Faculty of Veterinary Medicine and Animal Science, Poznan University of Life Sciences, Wojska Polskiego 71C, 60-625 Poznan, Poland;
| | | |
Collapse
|
3
|
Dunn P, Annamdevula NS, Leavesley SJ, Rich TC, Phan AV. A two-dimensional finite element model of intercellular cAMP signaling through gap junction channels. J Biomech 2023; 152:111588. [PMID: 37094384 PMCID: PMC10173664 DOI: 10.1016/j.jbiomech.2023.111588] [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: 09/16/2022] [Revised: 04/02/2023] [Accepted: 04/11/2023] [Indexed: 04/26/2023]
Abstract
While cyclic adenosine monophosphate (cAMP) is typically considered an intracellular signal, it has been shown to spread between adjacent cells through connexin-based gap junction channels, promoting gap junctional intercellular communication (GJIC). Gap junction-mediated signaling is critical for the coordinated function of many tissues, and have been linked with cardiovascular disease, neurogenerative disease, and cancers. In particular, it plays a complex role in tumor suppression or promotion. This work introduces a two-dimensional finite element model that can describe intercellular cAMP signaling in the presence of gap junctions on membrane interfaces. The model was utilized to simulate cAMP transfer through one and two gap junction channels on the interface of a cluster of two pulmonary microvascular endothelial cells. The simulation results were found to generally agree with what has been observed in the literature in terms of GJIC. The research outcomes suggest that the proposed model can be employed to evaluate the permeability properties of a gap junction channel if its cAMP volumetric flow rate can be experimentally measured.
Collapse
Affiliation(s)
- P Dunn
- Department of Mechanical, Aerospace and Biomedical Engineering, University of South Alabama, Mobile, AL 36688, USA
| | - N S Annamdevula
- Center for Lung Biology & Department of Pharmacology University of South Alabama, Mobile, AL 36688, USA
| | - S J Leavesley
- Center for Lung Biology & Department of Pharmacology University of South Alabama, Mobile, AL 36688, USA; Department of Chemical and Biomolecular Engineering, University of South Alabama, Mobile, AL 36688, USA
| | - T C Rich
- Center for Lung Biology & Department of Pharmacology University of South Alabama, Mobile, AL 36688, USA
| | - A-V Phan
- Department of Mechanical, Aerospace and Biomedical Engineering, University of South Alabama, Mobile, AL 36688, USA.
| |
Collapse
|
4
|
Chen P, Wu W, Zhang J, Chen J, Li Y, Sun L, Hou S, Yang J. Pathological mechanisms of connexin26-related hearing loss: Potassium recycling, ATP-calcium signaling, or energy supply? Front Mol Neurosci 2022; 15:976388. [PMID: 36187349 PMCID: PMC9520402 DOI: 10.3389/fnmol.2022.976388] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/11/2022] [Indexed: 11/17/2022] Open
Abstract
Hereditary deafness is one of the most common human birth defects. GJB2 gene mutation is the most genetic etiology. Gap junction protein 26 (connexin26, Cx26) encoded by the GJB2 gene, which is responsible for intercellular substance transfer and signal communication, plays a critical role in hearing acquisition and maintenance. The auditory character of different Connexin26 transgenic mice models can be classified into two types: profound congenital deafness and late-onset progressive hearing loss. Recent studies demonstrated that there are pathological changes including endocochlear potential reduction, active cochlear amplification impairment, cochlear developmental disorders, and so on, in connexin26 deficiency mice. Here, this review summarizes three main hypotheses to explain pathological mechanisms of connexin26-related hearing loss: potassium recycling disruption, adenosine-triphosphate-calcium signaling propagation disruption, and energy supply dysfunction. Elucidating pathological mechanisms underlying connexin26-related hearing loss can help develop new protective and therapeutic strategies for this common deafness. It is worthy of further study on the detailed cellular and molecular upstream mechanisms to modify connexin (channel) function.
Collapse
Affiliation(s)
- Penghui Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Wenjin Wu
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Jifang Zhang
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Junmin Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Yue Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Lianhua Sun
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Shule Hou
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
- *Correspondence: Shule Hou,
| | - Jun Yang
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
- Jun Yang,
| |
Collapse
|
5
|
Kraujalis T, Gudaitis L, Kraujaliene L, Snipas M, Palacios-Prado N, Verselis VK. The Amino Terminal Domain and Modulation of Connexin36 Gap Junction Channels by Intracellular Magnesium Ions. Front Physiol 2022; 13:839223. [PMID: 35264979 PMCID: PMC8899287 DOI: 10.3389/fphys.2022.839223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 01/28/2022] [Indexed: 11/13/2022] Open
Abstract
Electrical synapses between neurons in the mammalian CNS are predominantly formed of the connexin36 (Cx36) gap junction (GJ) channel protein. Unique among GJs formed of a number of other members of the Cx gene family, Cx36 GJs possess a high sensitivity to intracellular Mg2+ that can robustly act to modulate the strength of electrical synaptic transmission. Although a putative Mg2+ binding site was previously identified to reside in the aqueous pore in the first extracellular (E1) loop domain, the involvement of the N-terminal (NT) domain in the atypical response of Cx36 GJs to pH was shown to depend on intracellular levels of Mg2+. In this study, we examined the impact of amino acid substitutions in the NT domain on Mg2+ modulation of Cx36 GJs, focusing on positions predicted to line the pore funnel, which constitutes the cytoplasmic entrance of the channel pore. We find that charge substitutions at the 8th, 13th, and 18th positions had pronounced effects on Mg2+ sensitivity, particularly at position 13 at which an A13K substitution completely abolished sensitivity to Mg2+. To assess potential mechanisms of Mg2+ action, we constructed and tested a series of mathematical models that took into account gating of the component hemichannels in a Cx36 GJ channel as well as Mg2+ binding to each hemichannel in open and/or closed states. Simultaneous model fitting of measurements of junctional conductance, gj, and transjunctional Mg2+ fluxes using a fluorescent Mg2+ indicator suggested that the most viable mechanism for Cx36 regulation by Mg2+ entails the binding of Mg2+ to and subsequent stabilization of the closed state in each hemichannel. Reduced permeability to Mg2+ was also evident, particularly for the A13K substitution, but homology modeling of all charge-substituted NT variants showed only a moderate correlation between a reduction in the negative electrostatic potential and a reduction in the permeability to Mg2+ ions. Given the reported role of the E1 domain in Mg2+ binding together with the impact of NT substitutions on gating and the apparent state-dependence of Mg2+ binding, this study suggests that the NT domain can be an integral part of Mg2+ modulation of Cx36 GJs likely through the coupling of conformational changes between NT and E1 domains.
Collapse
Affiliation(s)
- Tadas Kraujalis
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Department of Applied Informatics, Kaunas University of Technology, Kaunas, Lithuania
- *Correspondence: Tadas Kraujalis,
| | - Lukas Gudaitis
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Lina Kraujaliene
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Mindaugas Snipas
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Department of Mathematical Modelling, Kaunas University of Technology, Kaunas, Lithuania
| | - Nicolás Palacios-Prado
- Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaiso, Valparaíso, Chile
| | - Vytas K. Verselis
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
6
|
Function and Plasticity of Electrical Synapses in the Mammalian Brain: Role of Non-Junctional Mechanisms. BIOLOGY 2022; 11:biology11010081. [PMID: 35053079 PMCID: PMC8773336 DOI: 10.3390/biology11010081] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 01/27/2023]
Abstract
Simple Summary Relevant brain functions, such as perception, organization of behavior, and cognitive processes, are the outcome of information processing by neural circuits. Within these circuits, communication between neurons mainly relies on two modalities of synaptic transmission: chemical and electrical. Moreover, changes in the strength of these connections, aka synaptic plasticity, are believed to underlie processes of learning and memory, and its dysfunction has been suggested to underlie a variety of neurological disorders. While the relevance of chemical transmission and its plastic changes are known in great detail, analogous mechanisms and functional impact of their electrical counterparts were only recently acknowledged. In this article, we review the basic physical principles behind electrical transmission between neurons, the plethora of functional operations supported by this modality of neuron-to-neuron communication, as well as the basic principles of plasticity at these synapses. Abstract Electrical transmission between neurons is largely mediated by gap junctions. These junctions allow the direct flow of electric current between neurons, and in mammals, they are mostly composed of the protein connexin36. Circuits of electrically coupled neurons are widespread in these animals. Plus, experimental and theoretical evidence supports the notion that, beyond synchronicity, these circuits are able to perform sophisticated operations such as lateral excitation and inhibition, noise reduction, as well as the ability to selectively respond upon coincident excitatory inputs. Although once considered stereotyped and unmodifiable, we now know that electrical synapses are subject to modulation and, by reconfiguring neural circuits, these modulations can alter relevant operations. The strength of electrical synapses depends on the gap junction resistance, as well as on its functional interaction with the electrophysiological properties of coupled neurons. In particular, voltage and ligand gated channels of the non-synaptic membrane critically determine the efficacy of transmission at these contacts. Consistently, modulatory actions on these channels have been shown to represent relevant mechanisms of plasticity of electrical synaptic transmission. Here, we review recent evidence on the regulation of electrical synapses of mammals, the underlying molecular mechanisms, and the possible ways in which they affect circuit function.
Collapse
|
7
|
Ngezahayo A, Ruhe FA. Connexins in the development and physiology of stem cells. Tissue Barriers 2021; 9:1949242. [PMID: 34227910 DOI: 10.1080/21688370.2021.1949242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Connexins (Cxs) form gap junction (GJ) channels linking vertebrate cells. During embryogenesis, Cxs are expressed as early as the 4-8 cell stage. As cells differentiate into pluripotent stem cells (PSCs) and during gastrulation, the Cx expression pattern is adapted. Knockdown of Cx43 and Cx45 does not interfere with embryogenic development until the blastula stage, questioning the role of Cxs in PSC physiology and development. Studies in cultivated and induced PSCs (iPSCs) showed that Cx43 is essential for the maintenance of self-renewal and the expression of pluripotency markers. It was found that the role of Cxs in PSCs is more related to regulation of transcription or cell-cell adherence than to formation of GJ channels. Furthermore, a crucial role of Cxs for the self-renewal and differentiation was shown in cultivated adult mesenchymal stem cells. This review aims to highlight aspects that link Cxs to the function and physiology of stem cell development.
Collapse
Affiliation(s)
- Anaclet Ngezahayo
- Dept. Cell Physiology and Biophysics, Institute of Cell Biology and Biophysics, Leibniz University Hannover, Hannover, Germany.,Center for Systems Neuroscience (ZSN), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Frederike A Ruhe
- Dept. Cell Physiology and Biophysics, Institute of Cell Biology and Biophysics, Leibniz University Hannover, Hannover, Germany
| |
Collapse
|
8
|
Connexins in the Heart: Regulation, Function and Involvement in Cardiac Disease. Int J Mol Sci 2021; 22:ijms22094413. [PMID: 33922534 PMCID: PMC8122935 DOI: 10.3390/ijms22094413] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/12/2021] [Accepted: 04/20/2021] [Indexed: 12/20/2022] Open
Abstract
Connexins are a family of transmembrane proteins that play a key role in cardiac physiology. Gap junctional channels put into contact the cytoplasms of connected cardiomyocytes, allowing the existence of electrical coupling. However, in addition to this fundamental role, connexins are also involved in cardiomyocyte death and survival. Thus, chemical coupling through gap junctions plays a key role in the spreading of injury between connected cells. Moreover, in addition to their involvement in cell-to-cell communication, mounting evidence indicates that connexins have additional gap junction-independent functions. Opening of unopposed hemichannels, located at the lateral surface of cardiomyocytes, may compromise cell homeostasis and may be involved in ischemia/reperfusion injury. In addition, connexins located at non-canonical cell structures, including mitochondria and the nucleus, have been demonstrated to be involved in cardioprotection and in regulation of cell growth and differentiation. In this review, we will provide, first, an overview on connexin biology, including their synthesis and degradation, their regulation and their interactions. Then, we will conduct an in-depth examination of the role of connexins in cardiac pathophysiology, including new findings regarding their involvement in myocardial ischemia/reperfusion injury, cardiac fibrosis, gene transcription or signaling regulation.
Collapse
|
9
|
Stephan J, Eitelmann S, Zhou M. Approaches to Study Gap Junctional Coupling. Front Cell Neurosci 2021; 15:640406. [PMID: 33776652 PMCID: PMC7987795 DOI: 10.3389/fncel.2021.640406] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/03/2021] [Indexed: 12/17/2022] Open
Abstract
Astrocytes and oligodendrocytes are main players in the brain to ensure ion and neurotransmitter homeostasis, metabolic supply, and fast action potential propagation in axons. These functions are fostered by the formation of large syncytia in which mainly astrocytes and oligodendrocytes are directly coupled. Panglial networks constitute on connexin-based gap junctions in the membranes of neighboring cells that allow the passage of ions, metabolites, and currents. However, these networks are not uniform but exhibit a brain region-dependent heterogeneous connectivity influencing electrical communication and intercellular ion spread. Here, we describe different approaches to analyze gap junctional communication in acute tissue slices that can be implemented easily in most electrophysiology and imaging laboratories. These approaches include paired recordings, determination of syncytial isopotentiality, tracer coupling followed by analysis of network topography, and wide field imaging of ion sensitive dyes. These approaches are capable to reveal cellular heterogeneity causing electrical isolation of functional circuits, reduced ion-transfer between different cell types, and anisotropy of tracer coupling. With a selective or combinatory use of these methods, the results will shed light on cellular properties of glial cells and their contribution to neuronal function.
Collapse
Affiliation(s)
- Jonathan Stephan
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sara Eitelmann
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Min Zhou
- Department of Neuroscience, Wexner Medical Center, Ohio State University, Columbus, OH, United States
| |
Collapse
|
10
|
A Cellular Assay for the Identification and Characterization of Connexin Gap Junction Modulators. Int J Mol Sci 2021; 22:ijms22031417. [PMID: 33572565 PMCID: PMC7866863 DOI: 10.3390/ijms22031417] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 12/18/2022] Open
Abstract
Connexin gap junctions (Cx GJs) enable the passage of small molecules and ions between cells and are therefore important for cell-to-cell communication. Their dysfunction is associated with diseases, and small molecules acting as modulators of GJs may therefore be useful as therapeutic drugs. To identify GJ modulators, suitable assays are needed that allow compound screening. In the present study, we established a novel assay utilizing HeLa cells recombinantly expressing Cx43. Donor cells additionally expressing the Gs protein-coupled adenosine A2A receptor, and biosensor cells expressing a cAMP-sensitive GloSensor luciferase were established. Adenosine A2A receptor activation in the donor cells using a selective agonist results in intracellular cAMP production. The negatively charged cAMP migrates via the Cx43 gap junctions to the biosensor cells and can there be measured by the cAMP-dependent luminescence signal. Cx43 GJ modulators can be expected to impact the transfer of cAMP from the donor to the biosensor cells, since cAMP transit is only possible via GJs. The new assay was validated by testing the standard GJ inhibitor carbenoxolon, which showed a concentration-dependent inhibition of the signal and an IC50 value that was consistent with previously reported values. The assay was demonstrated to be suitable for high-throughput screening.
Collapse
|
11
|
Connexins and cAMP Cross-Talk in Cancer Progression and Metastasis. Cancers (Basel) 2020; 13:cancers13010058. [PMID: 33379194 PMCID: PMC7795795 DOI: 10.3390/cancers13010058] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/21/2020] [Accepted: 12/21/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Different connexins play diverse roles in cancers, either tumor-suppressing or tumor-promoting. In lung cancer, Cx43 serves as a tumor suppressor at the early stage, but it can also be a tumor-promotor at an advanced stage and during metastasis. Moreover, other connexins, including Cx26, Cx31.1, and Cx32, can be tumor suppressors. In contrast, Cx30.3 can be a tumor-promotor. The roles of different connexins in different cancers have also been established. Cx43 acts as a tumor suppressor in colorectal cancer, breast cancer, and glioma, whereas Cx32 can be a suppressor in liver tumors and hepatocarcinogenesis. Cx26 can be a tumor suppressor in mammary tumors; in contrast, it can be a tumor-promotor in melanoma. Existing drugs/molecules targeting the cAMP/PKA/connexin axis act to regulate channel opening/closing. Mimic peptides, such as Gap19, Gap26, and Gap 27 block hemichannels, mimetic peptides, and CT9/CT10 and promote hemichannel opening and also hemichannel closing. Abstract Connexin-containing gap junctions mediate the direct exchange of small molecules between cells, thus promoting cell–cell communication. Connexins (Cxs) have been widely studied as key tumor-suppressors. However, certain Cx subtypes, such as Cx43 and Cx26, are overexpressed in metastatic tumor lesions. Cyclic adenosine monophosphate (cAMP) signaling regulates Cx expression and function via transcriptional control and phosphorylation. cAMP also passes through gap junction channels between adjacent cells, regulating cell cycle progression, particularly in cancer cell populations. Low levels of cAMP are sufficient to activate key effectors. The present review evaluates the mechanisms underlying Cx regulation by cAMP signaling and the role of gap junctions in cancer progression and metastasis. A deeper understanding of these processes might facilitate the development of novel anticancer drugs.
Collapse
|
12
|
Rose J, Brian C, Pappa A, Panayiotidis MI, Franco R. Mitochondrial Metabolism in Astrocytes Regulates Brain Bioenergetics, Neurotransmission and Redox Balance. Front Neurosci 2020; 14:536682. [PMID: 33224019 PMCID: PMC7674659 DOI: 10.3389/fnins.2020.536682] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 10/14/2020] [Indexed: 01/17/2023] Open
Abstract
In the brain, mitochondrial metabolism has been largely associated with energy production, and its dysfunction is linked to neuronal cell loss. However, the functional role of mitochondria in glial cells has been poorly studied. Recent reports have demonstrated unequivocally that astrocytes do not require mitochondria to meet their bioenergetics demands. Then, the question remaining is, what is the functional role of mitochondria in astrocytes? In this work, we review current evidence demonstrating that mitochondrial central carbon metabolism in astrocytes regulates overall brain bioenergetics, neurotransmitter homeostasis and redox balance. Emphasis is placed in detailing carbon source utilization (glucose and fatty acids), anaplerotic inputs and cataplerotic outputs, as well as carbon shuttles to neurons, which highlight the metabolic specialization of astrocytic mitochondria and its relevance to brain function.
Collapse
Affiliation(s)
- Jordan Rose
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE, United States.,School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Christian Brian
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE, United States.,School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Aglaia Pappa
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - Mihalis I Panayiotidis
- Department of Electron Microscopy & Molecular Pathology, Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus
| | - Rodrigo Franco
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE, United States.,School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| |
Collapse
|
13
|
Lagos-Cabré R, Burgos-Bravo F, Avalos AM, Leyton L. Connexins in Astrocyte Migration. Front Pharmacol 2020; 10:1546. [PMID: 32009957 PMCID: PMC6974553 DOI: 10.3389/fphar.2019.01546] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/29/2019] [Indexed: 12/18/2022] Open
Abstract
Astrocytes have long been considered the supportive cells of the central nervous system, but during the last decades, they have gained much more attention because of their active participation in the modulation of neuronal function. For example, after brain damage, astrocytes become reactive and undergo characteristic morphological and molecular changes, such as hypertrophy and increase in the expression of glial fibrillary acidic protein (GFAP), in a process known as astrogliosis. After severe damage, astrocytes migrate to the lesion site and proliferate, which leads to the formation of a glial scar. At this scar-forming stage, astrocytes secrete many factors, such as extracellular matrix proteins, cytokines, growth factors and chondroitin sulfate proteoglycans, stop migrating, and the process is irreversible. Although reactive gliosis is a normal physiological response that can protect brain cells from further damage, it also has detrimental effects on neuronal survival, by creating a hostile and non-permissive environment for axonal repair. The transformation of astrocytes from reactive to scar-forming astrocytes highlights migration as a relevant regulator of glial scar formation, and further emphasizes the importance of efficient communication between astrocytes in order to orchestrate cell migration. The coordination between astrocytes occurs mainly through Connexin (Cx) channels, in the form of direct cell-cell contact (gap junctions, GJs) or contact between the extracellular matrix and the astrocytes (hemichannels, HCs). Reactive astrocytes increase the expression levels of several proteins involved in astrocyte migration, such as αvβ3 Integrin, Syndecan-4 proteoglycan, the purinergic receptor P2X7, Pannexin1, and Cx43 HCs. Evidence has indicated that Cx43 HCs play a role in regulating astrocyte migration through the release of small molecules to the extracellular space, which then activate receptors in the same or adjacent cells to continue the signaling cascades required for astrocyte migration. In this review, we describe the communication of astrocytes through Cxs, the role of Cxs in inflammation and astrocyte migration, and discuss the molecular mechanisms that regulate Cx43 HCs, which may provide a therapeutic window of opportunity to control astrogliosis and the progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Raúl Lagos-Cabré
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Center for Studies on Exercise, Metabolism and Cancer (CEMC), Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Francesca Burgos-Bravo
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Center for Studies on Exercise, Metabolism and Cancer (CEMC), Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Ana María Avalos
- Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Center for Studies on Exercise, Metabolism and Cancer (CEMC), Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| |
Collapse
|
14
|
Chien J, Wolf FW, Grosche S, Yosef N, Garriga G, Mörck C. The Enigmatic Canal-Associated Neurons Regulate Caenorhabditis elegans Larval Development Through a cAMP Signaling Pathway. Genetics 2019; 213:1465-1478. [PMID: 31619445 PMCID: PMC6893374 DOI: 10.1534/genetics.119.302628] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 10/13/2019] [Indexed: 11/18/2022] Open
Abstract
Caenorhabditis elegans larval development requires the function of the two Canal-Associated Neurons (CANs): killing the CANs by laser microsurgery or disrupting their development by mutating the gene ceh-10 results in early larval arrest. How these cells promote larval development, however, remains a mystery. In screens for mutations that bypass CAN function, we identified the gene kin-29, which encodes a member of the Salt-Inducible Kinase (SIK) family and a component of a conserved pathway that regulates various C. elegans phenotypes. Like kin-29 loss, gain-of-function mutations in genes that may act upstream of kin-29 or growth in cyclic-AMP analogs bypassed ceh-10 larval arrest, suggesting that a conserved adenylyl cyclase/PKA pathway inhibits KIN-29 to promote larval development, and that loss of CAN function results in dysregulation of KIN-29 and larval arrest. The adenylyl cyclase ACY-2 mediates CAN-dependent larval development: acy-2 mutant larvae arrested development with a similar phenotype to ceh-10 mutants, and the arrest phenotype was suppressed by mutations in kin-29 ACY-2 is expressed predominantly in the CANs, and we provide evidence that the acy-2 functions in the CANs to promote larval development. By contrast, cell-specific expression experiments suggest that kin-29 acts in both the hypodermis and neurons, but not in the CANs. Based on our findings, we propose two models for how ACY-2 activity in the CANs regulates KIN-29 in target cells.
Collapse
Affiliation(s)
- Jason Chien
- Department of Chemistry and Molecular Biology, University of Gothenburg, Sweden 405 30
| | - Fred W Wolf
- Department of Molecular and Cell Biology, University of California, Merced, California 95343
| | - Sarah Grosche
- Department of Chemistry and Molecular Biology, University of Gothenburg, Sweden 405 30
| | - Nebeyu Yosef
- Department of Chemistry and Molecular Biology, University of Gothenburg, Sweden 405 30
| | - Gian Garriga
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720-3204
| | - Catarina Mörck
- Department of Chemistry and Molecular Biology, University of Gothenburg, Sweden 405 30
| |
Collapse
|
15
|
Mammano F. Inner Ear Connexin Channels: Roles in Development and Maintenance of Cochlear Function. Cold Spring Harb Perspect Med 2019; 9:a033233. [PMID: 30181354 PMCID: PMC6601451 DOI: 10.1101/cshperspect.a033233] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Connexin 26 and connexin 30 are the prevailing isoforms in the epithelial and connective tissue gap junction systems of the developing and mature cochlea. The most frequently encountered variants of the genes that encode these connexins, which are transcriptionally coregulated, determine complete loss of protein function and are the predominant cause of prelingual hereditary deafness. Reducing connexin 26 expression by Cre/loxP recombination in the inner ear of adult mice results in a decreased endocochlear potential, increased hearing thresholds, and loss of >90% of outer hair cells, indicating that this connexin is essential for maintenance of cochlear function. In the developing cochlea, connexins are necessary for intercellular calcium signaling activity. Ribbon synapses and basolateral membrane currents fail to mature in inner hair cells of mice that are born with reduced connexin expression, even though hair cells do not express any connexin. In contrast, pannexin 1, an alternative mediator of intercellular signaling, is dispensable for hearing acquisition and auditory function.
Collapse
Affiliation(s)
- Fabio Mammano
- University of Padova, Department of Physics and Astronomy "G. Galilei," Padova 35129, Italy
- CNR Institute of Cell Biology and Neurobiology, Monterotondo 00015, Italy
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
16
|
Schadzek P, Stahl Y, Preller M, Ngezahayo A. Analysis of the dominant mutation N188T of human connexin46 (hCx46) using concatenation and molecular dynamics simulation. FEBS Open Bio 2019; 9:840-850. [PMID: 31034164 PMCID: PMC6487695 DOI: 10.1002/2211-5463.12624] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/06/2019] [Accepted: 02/26/2019] [Indexed: 11/27/2022] Open
Abstract
Connexins (Cx) are proteins that form cell-to-cell gap junction channels. A mutation at position 188 in the second extracellular loop (E2) domain of hCx46 has been linked to an autosomal dominant zonular pulverulent cataract. As it is dominantly inherited, it is possible that the mutant variant affects the co-expressed wild-type Cx and/or its interaction with other cellular components. Here, we proposed to use concatenated hCx46wt-hCx46N188T and hCx46N188T-hCx46wt to analyze how hCx46N188T affected co-expressed hCx46wt to achieve a dominant inheritance. Heterodimer hCx46wt-hCx46N188T formed fewer gap junction plaques compared to homodimer hCx46wt-hCx46wt, while the hCx46N188T-hCx46N188T homodimer formed almost no gap junction plaques. Dye uptake experiments showed that hemichannels of concatenated variants were similar to hemichannels of monomers. Molecular dynamics simulations revealed that for docking, the N188 of a protomer was engaged in hydrogen bonds (HBs) with R180, N189, and D191 of the counterpart protomer of the adjacent hemichannel. T188 suppressed the formation of HBs between protomers. Molecular dynamics simulations of an equimolar hCx46wt/hCx46N188T gap junction channel revealed a reduced number of HBs between protomers, suggesting reduction of gap junction channels between lens fibers co-expressing the variants.
Collapse
Affiliation(s)
- Patrik Schadzek
- Institute of Cell Biology and BiophysicsDepartment of Cell Physiology and BiophysicsLeibniz University HannoverGermany
| | - Yannick Stahl
- Institute of Cell Biology and BiophysicsDepartment of Cell Physiology and BiophysicsLeibniz University HannoverGermany
| | - Matthias Preller
- Institute for Biophysical ChemistryHannover Medical School (MHH)Germany
- Centre for Structural Systems Biology, DESY‐CampusHamburgGermany
| | - Anaclet Ngezahayo
- Institute of Cell Biology and BiophysicsDepartment of Cell Physiology and BiophysicsLeibniz University HannoverGermany
- Center for System Neurosciences (ZSN)HannoverGermany
| |
Collapse
|
17
|
Wadle SL, Augustin V, Langer J, Jabs R, Philippot C, Weingarten DJ, Rose CR, Steinhäuser C, Stephan J. Anisotropic Panglial Coupling Reflects Tonotopic Organization in the Inferior Colliculus. Front Cell Neurosci 2018; 12:431. [PMID: 30542265 PMCID: PMC6277822 DOI: 10.3389/fncel.2018.00431] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 10/31/2018] [Indexed: 12/17/2022] Open
Abstract
Astrocytes and oligodendrocytes in different brain regions form panglial networks and the topography of such networks can correlate with neuronal topography and function. Astrocyte-oligodendrocyte networks in the lateral superior olive (LSO)-an auditory brainstem nucleus-were found to be anisotropic with a preferred orientation orthogonally to the tonotopic axis. We hypothesized that such a specialization might be present in other tonotopically organized brainstem nuclei, too. Thus, we analyzed gap junctional coupling in the center of the inferior colliculus (IC)-another nucleus of the auditory brainstem that exhibits tonotopic organization. In acute brainstem slices obtained from mice, IC networks were traced employing whole-cell patch-clamp recordings of single sulforhodamine (SR) 101-identified astrocytes and concomitant intracellular loading of the gap junction-permeable tracer neurobiotin. The majority of dye-coupled networks exhibited an oval topography, which was preferentially oriented orthogonal to the tonotopic axis. Astrocyte processes showed preferentially the same orientation indicating a correlation between astrocyte and network topography. In addition to SR101-positive astrocytes, IC networks contained oligodendrocytes. Using Na+ imaging, we analyzed the capability of IC networks to redistribute small ions. Na+ bi-directionally diffused between SR101-positive astrocytes and SR101-negative cells-presumably oligodendrocytes-showing the functionality of IC networks. Taken together, our results demonstrate that IC astrocytes and IC oligodendrocytes form functional anisotropic panglial networks that are preferentially oriented orthogonal to the tonotopic axis. Thus, our data indicate that the topographic specialization of glial networks seen in IC and LSO might be a general feature of tonotopically organized auditory brainstem nuclei.
Collapse
Affiliation(s)
- Simon L Wadle
- Animal Physiology Group, Department of Biology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Vanessa Augustin
- Animal Physiology Group, Department of Biology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Julia Langer
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Ronald Jabs
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Camille Philippot
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Dennis J Weingarten
- Animal Physiology Group, Department of Biology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Christine R Rose
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Jonathan Stephan
- Animal Physiology Group, Department of Biology, University of Kaiserslautern, Kaiserslautern, Germany
| |
Collapse
|
18
|
Schadzek P, Hermes D, Stahl Y, Dilger N, Ngezahayo A. Concatenation of Human Connexin26 (hCx26) and Human Connexin46 (hCx46) for the Analysis of Heteromeric Gap Junction Hemichannels and Heterotypic Gap Junction Channels. Int J Mol Sci 2018; 19:E2742. [PMID: 30217016 PMCID: PMC6163895 DOI: 10.3390/ijms19092742] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/10/2018] [Accepted: 09/11/2018] [Indexed: 12/16/2022] Open
Abstract
Gap junction channels and hemichannels formed by concatenated connexins were analyzed. Monomeric (hCx26, hCx46), homodimeric (hCx46-hCx46, hCx26-hCx26), and heterodimeric (hCx26-hCx46, hCx46-hCx26) constructs, coupled to GFP, were expressed in HeLa cells. Confocal microscopy showed that the tandems formed gap junction plaques with a reduced plaque area compared to monomeric hCx26 or hCx46. Dye transfer experiments showed that concatenation allows metabolic transfer. Expressed in Xenopus oocytes, the inside-out patch-clamp configuration showed single channels with a conductance of about 46 pS and 39 pS for hemichannels composed of hCx46 and hCx26 monomers, respectively, when chloride was replaced by gluconate on both membrane sides. The conductance was reduced for hCx46-hCx46 and hCx26-hCx26 homodimers, probably due to the concatenation. Heteromerized hemichannels, depending on the connexin-order, were characterized by substates at 26 pS and 16 pS for hCx46-hCx26 and 31 pS and 20 pS for hCx26-hCx46. Because of the linker between the connexins, the properties of the formed hemichannels and gap junction channels (e.g., single channel conductance) may not represent the properties of hetero-oligomerized channels. However, should the removal of the linker be successful, this method could be used to analyze the electrical and metabolic selectivity of such channels and the physiological consequences for a tissue.
Collapse
Affiliation(s)
- Patrik Schadzek
- Institut für Biophysik, Leibniz Universität Hannover, Herrenhäuser Straße 2, 30419 Hannover, Germany.
| | - Doris Hermes
- Institut für Biophysik, Leibniz Universität Hannover, Herrenhäuser Straße 2, 30419 Hannover, Germany.
- Department of Clinical Neurophysiology, University of Göttingen, Robert-Koch Str. 40, D-37075 Göttingen, Germany.
| | - Yannick Stahl
- Institut für Biophysik, Leibniz Universität Hannover, Herrenhäuser Straße 2, 30419 Hannover, Germany.
| | - Nadine Dilger
- Institut für Biophysik, Leibniz Universität Hannover, Herrenhäuser Straße 2, 30419 Hannover, Germany.
| | - Anaclet Ngezahayo
- Institut für Biophysik, Leibniz Universität Hannover, Herrenhäuser Straße 2, 30419 Hannover, Germany.
- Zentrum für Systemische Neurowissenschaften Stiftung Tierärztliche Hochschule Hannover, Bünteweg 2, 30559 Hannover, Germany.
| |
Collapse
|
19
|
Rimkute L, Kraujalis T, Snipas M, Palacios-Prado N, Jotautis V, Skeberdis VA, Bukauskas FF. Modulation of Connexin-36 Gap Junction Channels by Intracellular pH and Magnesium Ions. Front Physiol 2018; 9:362. [PMID: 29706896 PMCID: PMC5906587 DOI: 10.3389/fphys.2018.00362] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 03/23/2018] [Indexed: 11/13/2022] Open
Abstract
Connexin-36 (Cx36) protein forms gap junction (GJ) channels in pancreatic beta cells and is also the main Cx isoform forming electrical synapses in the adult mammalian brain. Cx36 GJs can be regulated by intracellular pH (pHi) and cytosolic magnesium ion concentration ([Mg2+]i), which can vary significantly under various physiological and pathological conditions. However, the combined effect and relationship of these two factors over Cx36-dependent coupling have not been previously studied in detail. Our experimental results in HeLa cells expressing Cx36 show that changes in both pHi and [Mg2+]i affect junctional conductance (gj) in an interdependent manner; in other words, intracellular acidification cause increase or decay in gj depending on whether [Mg2+]i is high or low, respectively, and intracellular alkalization cause reduction in gj independently of [Mg2+]i. Our experimental and modelling data support the hypothesis that Cx36 GJ channels contain two separate gating mechanisms, and both are differentially sensitive to changes in pHi and [Mg2+]i. Using recombinant Cx36 we found that two glutamate residues in the N-terminus could be partly responsible for the observed interrelated effect of pHi and [Mg2+]i. Mutation of glutamate at position 8 attenuated the stimulatory effect of intracellular acidification at high [Mg2+]i, while mutation at position 12 and double mutation at both positions reversed stimulatory effect to inhibition. Moreover, Cx36*E8Q lost the initial increase of gj at low [Mg2+]i and double mutation lost the sensitivity to high [Mg2+]i. These results suggest that E8 and E12 are involved in regulation of Cx36 GJ channels by Mg2+ and H+ ions.
Collapse
Affiliation(s)
- Lina Rimkute
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Tadas Kraujalis
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Department of Applied Informatics, Kaunas University of Technology, Kaunas, Lithuania
| | - Mindaugas Snipas
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Department of Mathematical Modelling, Kaunas University of Technology, Kaunas, Lithuania
| | - Nicolas Palacios-Prado
- Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Vaidas Jotautis
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Vytenis A. Skeberdis
- Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | |
Collapse
|
20
|
Hong K, Cope EL, DeLalio LJ, Marziano C, Isakson BE, Sonkusare SK. TRPV4 (Transient Receptor Potential Vanilloid 4) Channel-Dependent Negative Feedback Mechanism Regulates G q Protein-Coupled Receptor-Induced Vasoconstriction. Arterioscler Thromb Vasc Biol 2018; 38:542-554. [PMID: 29301784 DOI: 10.1161/atvbaha.117.310038] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 12/18/2017] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Several physiological stimuli activate smooth muscle cell (SMC) GqPCRs (Gq protein-coupled receptors) to cause vasoconstriction. As a protective mechanism against excessive vasoconstriction, SMC GqPCR stimulation invokes endothelial cell vasodilatory signaling. Whether Ca2+ influx in endothelial cells contributes to the regulation of GqPCR-induced vasoconstriction remains unknown. Ca2+ influx through TRPV4 (transient receptor potential vanilloid 4) channels is a key regulator of endothelium-dependent vasodilation. We hypothesized that SMC GqPCR stimulation engages endothelial TRPV4 channels to limit vasoconstriction. APPROACH AND RESULTS Using high-speed confocal microscopy to record unitary Ca2+ influx events through TRPV4 channels (TRPV4 sparklets), we report that activation of SMC α1ARs (alpha1-adrenergic receptors) with phenylephrine or thromboxane A2 receptors with U46619 stimulated TRPV4 sparklets in the native endothelium from mesenteric arteries. Activation of endothelial TRPV4 channels did not require an increase in Ca2+ as indicated by the lack of effect of L-type Ca2+ channel activator or chelator of intracellular Ca2+ EGTA-AM. However, gap junction communication between SMCs and endothelial cells was required for phenylephrine activation or U46619 activation of endothelial TRPV4 channels. Lowering inositol 1,4,5-trisphosphate levels with phospholipase C inhibitor or lithium chloride suppressed phenylephrine activation of endothelial TRPV4 sparklets. Moreover, uncaging inositol 1,4,5-trisphosphate profoundly increased TRPV4 sparklet activity. In pressurized arteries, phenylephrine-induced vasoconstriction was followed by a slow, TRPV4-dependent vasodilation, reflecting activation of negative regulatory mechanism. Consistent with these data, phenylephrine induced a significantly higher increase in blood pressure in TRPV4-/- mice. CONCLUSIONS These results demonstrate that SMC GqPCR stimulation triggers inositol 1,4,5-trisphosphate-dependent activation of endothelial TRPV4 channels to limit vasoconstriction.
Collapse
Affiliation(s)
- Kwangseok Hong
- From the Robert M. Berne Cardiovascular Research Center (K.H., E.L.C., L.J.D., C.M., B.E.I., S.K.S.), Department of Molecular Physiology and Biological Physics (C.M., B.E.I., S.K.S.), and Department of Pharmacology (L.J.D., S.K.S), University of Virginia-School of Medicine, Charlottesville
| | - Eric L Cope
- From the Robert M. Berne Cardiovascular Research Center (K.H., E.L.C., L.J.D., C.M., B.E.I., S.K.S.), Department of Molecular Physiology and Biological Physics (C.M., B.E.I., S.K.S.), and Department of Pharmacology (L.J.D., S.K.S), University of Virginia-School of Medicine, Charlottesville
| | - Leon J DeLalio
- From the Robert M. Berne Cardiovascular Research Center (K.H., E.L.C., L.J.D., C.M., B.E.I., S.K.S.), Department of Molecular Physiology and Biological Physics (C.M., B.E.I., S.K.S.), and Department of Pharmacology (L.J.D., S.K.S), University of Virginia-School of Medicine, Charlottesville
| | - Corina Marziano
- From the Robert M. Berne Cardiovascular Research Center (K.H., E.L.C., L.J.D., C.M., B.E.I., S.K.S.), Department of Molecular Physiology and Biological Physics (C.M., B.E.I., S.K.S.), and Department of Pharmacology (L.J.D., S.K.S), University of Virginia-School of Medicine, Charlottesville
| | - Brant E Isakson
- From the Robert M. Berne Cardiovascular Research Center (K.H., E.L.C., L.J.D., C.M., B.E.I., S.K.S.), Department of Molecular Physiology and Biological Physics (C.M., B.E.I., S.K.S.), and Department of Pharmacology (L.J.D., S.K.S), University of Virginia-School of Medicine, Charlottesville
| | - Swapnil K Sonkusare
- From the Robert M. Berne Cardiovascular Research Center (K.H., E.L.C., L.J.D., C.M., B.E.I., S.K.S.), Department of Molecular Physiology and Biological Physics (C.M., B.E.I., S.K.S.), and Department of Pharmacology (L.J.D., S.K.S), University of Virginia-School of Medicine, Charlottesville.
| |
Collapse
|
21
|
Leybaert L, Lampe PD, Dhein S, Kwak BR, Ferdinandy P, Beyer EC, Laird DW, Naus CC, Green CR, Schulz R. Connexins in Cardiovascular and Neurovascular Health and Disease: Pharmacological Implications. Pharmacol Rev 2017; 69:396-478. [PMID: 28931622 PMCID: PMC5612248 DOI: 10.1124/pr.115.012062] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Connexins are ubiquitous channel forming proteins that assemble as plasma membrane hemichannels and as intercellular gap junction channels that directly connect cells. In the heart, gap junction channels electrically connect myocytes and specialized conductive tissues to coordinate the atrial and ventricular contraction/relaxation cycles and pump function. In blood vessels, these channels facilitate long-distance endothelial cell communication, synchronize smooth muscle cell contraction, and support endothelial-smooth muscle cell communication. In the central nervous system they form cellular syncytia and coordinate neural function. Gap junction channels are normally open and hemichannels are normally closed, but pathologic conditions may restrict gap junction communication and promote hemichannel opening, thereby disturbing a delicate cellular communication balance. Until recently, most connexin-targeting agents exhibited little specificity and several off-target effects. Recent work with peptide-based approaches has demonstrated improved specificity and opened avenues for a more rational approach toward independently modulating the function of gap junctions and hemichannels. We here review the role of connexins and their channels in cardiovascular and neurovascular health and disease, focusing on crucial regulatory aspects and identification of potential targets to modify their function. We conclude that peptide-based investigations have raised several new opportunities for interfering with connexins and their channels that may soon allow preservation of gap junction communication, inhibition of hemichannel opening, and mitigation of inflammatory signaling.
Collapse
Affiliation(s)
- Luc Leybaert
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Paul D Lampe
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Stefan Dhein
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Brenda R Kwak
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Peter Ferdinandy
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Eric C Beyer
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Dale W Laird
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Christian C Naus
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Colin R Green
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Rainer Schulz
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| |
Collapse
|
22
|
Xu L, Carrer A, Zonta F, Qu Z, Ma P, Li S, Ceriani F, Buratto D, Crispino G, Zorzi V, Ziraldo G, Bruno F, Nardin C, Peres C, Mazzarda F, Salvatore AM, Raspa M, Scavizzi F, Chu Y, Xie S, Yang X, Liao J, Liu X, Wang W, Wang S, Yang G, Lerner RA, Mammano F. Design and Characterization of a Human Monoclonal Antibody that Modulates Mutant Connexin 26 Hemichannels Implicated in Deafness and Skin Disorders. Front Mol Neurosci 2017; 10:298. [PMID: 29018324 PMCID: PMC5615210 DOI: 10.3389/fnmol.2017.00298] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 09/05/2017] [Indexed: 12/21/2022] Open
Abstract
Background: Mutations leading to changes in properties, regulation, or expression of connexin-made channels have been implicated in 28 distinct human hereditary diseases. Eight of these result from variants of connexin 26 (Cx26), a protein critically involved in cell-cell signaling in the inner ear and skin. Lack of non-toxic drugs with defined mechanisms of action poses a serious obstacle to therapeutic interventions for diseases caused by mutant connexins. In particular, molecules that specifically modulate connexin hemichannel function without affecting gap junction channels are considered of primary importance for the study of connexin hemichannel role in physiological as well as pathological conditions. Monoclonal antibodies developed in the last three decades have become the most important class of therapeutic biologicals. Recombinant methods permit rapid selection and improvement of monoclonal antibodies from libraries with large diversity. Methods: By screening a combinatorial library of human single-chain fragment variable (scFv) antibodies expressed in phage, we identified a candidate that binds an extracellular epitope of Cx26. We characterized antibody action using a variety of biochemical and biophysical assays in HeLa cells, organotypic cultures of mouse cochlea and human keratinocyte-derived cells. Results: We determined that the antibody is a remarkably efficient, non-toxic, and completely reversible inhibitor of hemichannels formed by connexin 26 and does not affect direct cell-cell communication via gap junction channels. Importantly, we also demonstrate that the antibody efficiently inhibits hyperative mutant Cx26 hemichannels implicated in autosomal dominant non-syndromic hearing impairment accompanied by keratitis and hystrix-like ichthyosis-deafness (KID/HID) syndrome. We solved the crystal structure of the antibody, identified residues that are critical for binding and used molecular dynamics to uncover its mechanism of action. Conclusions: Although further studies will be necessary to validate the effect of the antibody in vivo, the methodology described here can be extended to select antibodies against hemichannels composed by other connexin isoforms and, consequently, to target other pathologies associated with hyperactive hemichannels. Our study highlights the potential of this approach and identifies connexins as therapeutic targets addressable by screening phage display libraries expressing human randomized antibodies.
Collapse
Affiliation(s)
- Liang Xu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China.,Institute of Biochemistry and Cell Biology, Shanghai Institute for Biological Sciences, Chinese Academy of SciencesShanghai, China
| | - Andrea Carrer
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Physics and Astronomy "G. Galilei,", University of PadovaPadova, Italy
| | - Francesco Zonta
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China.,CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy
| | - Zhihu Qu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Peixiang Ma
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Sheng Li
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Federico Ceriani
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Physics and Astronomy "G. Galilei,", University of PadovaPadova, Italy
| | - Damiano Buratto
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Physics and Astronomy "G. Galilei,", University of PadovaPadova, Italy
| | - Giulia Crispino
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Physics and Astronomy "G. Galilei,", University of PadovaPadova, Italy.,Venetian Institute of Molecular MedicinePadova, Italy
| | - Veronica Zorzi
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Institute of Otolaryngology, Catholic University School of MedicineRome, Italy
| | - Gaia Ziraldo
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Physics and Astronomy "G. Galilei,", University of PadovaPadova, Italy.,Institute of Otolaryngology, Catholic University School of MedicineRome, Italy
| | - Francesca Bruno
- Department of Physics and Astronomy "G. Galilei,", University of PadovaPadova, Italy.,Venetian Institute of Molecular MedicinePadova, Italy
| | - Chiara Nardin
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Science, Roma Tre UniversityRome, Italy
| | - Chiara Peres
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy
| | - Flavia Mazzarda
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Science, Roma Tre UniversityRome, Italy
| | - Anna M Salvatore
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy
| | - Marcello Raspa
- CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy
| | | | - Youjun Chu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Sichun Xie
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Xuemei Yang
- School of Life Science and Technology, Shanghai Tech UniversityShanghai, China
| | - Jun Liao
- School of Life Science and Technology, Shanghai Tech UniversityShanghai, China
| | - Xiao Liu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China.,Institute of Biochemistry and Cell Biology, Shanghai Institute for Biological Sciences, Chinese Academy of SciencesShanghai, China.,University of Chinese Academy of SciencesBeijing, China
| | - Wei Wang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Shanshan Wang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Guang Yang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China
| | - Richard A Lerner
- Department of Cell and Molecular Biology, The Scripps Research InstituteLa Jolla, CA, United States
| | - Fabio Mammano
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech UniversityShanghai, China.,CNR Institute of Cell Biology and NeurobiologyMonterotondo, Italy.,Department of Physics and Astronomy "G. Galilei,", University of PadovaPadova, Italy.,Venetian Institute of Molecular MedicinePadova, Italy
| |
Collapse
|
23
|
Kardiale „gap junctions“. ZEITSCHRIFT FUR HERZ THORAX UND GEFASSCHIRURGIE 2017. [DOI: 10.1007/s00398-016-0106-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
24
|
Mondal A, Sachse FB, Moreno AP. Modulation of Asymmetric Flux in Heterotypic Gap Junctions by Pore Shape, Particle Size and Charge. Front Physiol 2017; 8:206. [PMID: 28428758 PMCID: PMC5382223 DOI: 10.3389/fphys.2017.00206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/20/2017] [Indexed: 01/26/2023] Open
Abstract
Gap junction channels play a vital role in intercellular communication by connecting cytoplasm of adjoined cells through arrays of channel-pores formed at the common membrane junction. Their structure and properties vary depending on the connexin isoform(s) involved in forming the full gap junction channel. Lack of information on the molecular structure of gap junction channels has limited the development of computational tools for single channel studies. Currently, we rely on cumbersome experimental techniques that have limited capabilities. We have earlier reported a simplified Brownian dynamics gap junction pore model and demonstrated that variations in pore shape at the single channel level can explain some of the differences in permeability of heterotypic channels observed in in vitro experiments. Based on this computational model, we designed simulations to study the influence of pore shape, particle size and charge in homotypic and heterotypic pores. We simulated dye diffusion under whole cell voltage clamping. Our simulation studies with pore shape variations revealed a pore shape with maximal flux asymmetry in a heterotypic pore. We identified pore shape profiles that match the in silico flux asymmetry results to the in vitro results of homotypic and heterotypic gap junction formed out of Cx43 and Cx45. Our simulation results indicate that the channel's pore-shape established flux asymmetry and that flux asymmetry is primarily regulated by the sizes of the conical and/or cylindrical mouths at each end of the pore. Within the set range of particle size and charge, flux asymmetry was found to be independent of particle size and directly proportional to charge magnitude. While particle charge was vital to creating flux asymmetry, charge magnitude only scaled the observed flux asymmetry. Our studies identified the key factors that help predict asymmetry. Finally, we suggest the role of such flux asymmetry in creating concentration imbalances of messenger molecules in cardiomyocytes. We also assess the potency of fibroblasts in aggravating such imbalances through Cx43-Cx45 heterotypic channels in fibrotic heart tissue.
Collapse
Affiliation(s)
- Abhijit Mondal
- Department of Bioengineering, University of UtahSalt Lake City, UT, USA.,Nora Eccles Harrison Cardiovascular Research and Training Institute, University of UtahSalt Lake City, UT, USA
| | - Frank B Sachse
- Department of Bioengineering, University of UtahSalt Lake City, UT, USA.,Nora Eccles Harrison Cardiovascular Research and Training Institute, University of UtahSalt Lake City, UT, USA
| | - Alonso P Moreno
- Department of Bioengineering, University of UtahSalt Lake City, UT, USA.,Nora Eccles Harrison Cardiovascular Research and Training Institute, University of UtahSalt Lake City, UT, USA.,Department of Internal Medicine, Cardiology, University of UtahSalt Lake City, UT, USA
| |
Collapse
|
25
|
Zakkaroff C, Moore S, Dowding S, David T. 3D time-varying simulations of Ca 2+ dynamics in arterial coupled cells: A massively parallel implementation. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2017; 33:e02786. [PMID: 27062231 PMCID: PMC5298049 DOI: 10.1002/cnm.2786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 03/07/2016] [Accepted: 03/19/2016] [Indexed: 05/18/2023]
Abstract
Preferential locations of atherosclerotic plaque are strongly associated with the areas of low wall shear stress and disturbed haemodynamic characteristics such as flow detachment, flow recirculation and oscillatory flow. The areas of low wall shear stress are also associated with the reduced production of adenosine triphosphate in the endothelial layer, as well as the resulting reduced production of inositol trisphosphate (IP3 ). The subsequent variation in Ca2+ signalling and nitric oxide synthesis could lead to the impairment of the atheroprotective function played by nitric oxide. In previous studies, it has been suggested that the reduced IP3 and Ca2+ signalling can explain the correlation of atherosclerosis with induced low WSS and disturbed flow characteristics. The massively parallel implementation described in this article provides insight into the dynamics of coupled smooth muscle cells and endothelial cells mapped onto the surface of an idealised arterial bifurcation. We show that variations in coupling parameters, which model normal and pathological conditions, provide vastly different smooth muscle cell Ca2+ dynamics and wave propagation profiles. The extensibility of the coupled cells model and scalability of the implementation provide a solid framework for in silico investigations of the interaction between complex cellular chemistry and the macro-scale processes determined by fluid dynamics. © 2016 The Authors. International Journal for Numerical Methods in Biomedical Engineering published by John Wiley & Sons Ltd.
Collapse
Affiliation(s)
| | - Stephen Moore
- IBM Research Collaboratory for Life SciencesMelbourneAustralia
| | - Stewart Dowding
- UC HPC CentreUniversity of CanterburyChristchurchNew Zealand
| | - Tim David
- UC HPC CentreUniversity of CanterburyChristchurchNew Zealand
| |
Collapse
|
26
|
Klein M, Bopp T. Cyclic AMP Represents a Crucial Component of Treg Cell-Mediated Immune Regulation. Front Immunol 2016; 7:315. [PMID: 27621729 PMCID: PMC5002888 DOI: 10.3389/fimmu.2016.00315] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/02/2016] [Indexed: 11/13/2022] Open
Abstract
T regulatory (Treg) cells are one of the key players in the immune tolerance network, and a plethora of manuscripts have described their development and function in the course of the last two decades. Nevertheless, it is still a matter of debate as to which mechanisms and agents are employed by Treg cells, providing the basis of their suppressive potency. One of the important candidates is cyclic AMP (cAMP), which is long known as a potent suppressor at least of T cell activation and function. While this suppressive function by itself is widely accepted, the source and the mechanism of action of cAMP are less clear, and a multitude of seemingly contradictory data allow for, in principle, two different scenarios of cAMP-mediated suppression. In one scenario, Treg cells contain high amounts of cAMP and convey this small molecule via gap junction intercellular communication directly to the effector T cells (Teff) leading to their suppression. Alternatively, it was shown that Treg cells represent the origin of considerable amounts of adenosine, which trigger the adenylate cyclases in Teff cells via A2A and A2B receptors, thus strongly increasing intracellular cAMP. This review will present and discuss initial findings and recent developments concerning the function of cAMP for Treg cells and its impact on immune regulation.
Collapse
Affiliation(s)
- Matthias Klein
- University Medical Center, Institute for Immunology, Johannes Gutenberg-University , Mainz , Germany
| | - Tobias Bopp
- University Medical Center, Institute for Immunology, Johannes Gutenberg-University , Mainz , Germany
| |
Collapse
|
27
|
Krzyzanowski MC, Woldemariam S, Wood JF, Chaubey AH, Brueggemann C, Bowitch A, Bethke M, L’Etoile ND, Ferkey DM. Aversive Behavior in the Nematode C. elegans Is Modulated by cGMP and a Neuronal Gap Junction Network. PLoS Genet 2016; 12:e1006153. [PMID: 27459302 PMCID: PMC4961389 DOI: 10.1371/journal.pgen.1006153] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 06/08/2016] [Indexed: 01/03/2023] Open
Abstract
All animals rely on their ability to sense and respond to their environment to survive. However, the suitability of a behavioral response is context-dependent, and must reflect both an animal's life history and its present internal state. Based on the integration of these variables, an animal's needs can be prioritized to optimize survival strategies. Nociceptive sensory systems detect harmful stimuli and allow for the initiation of protective behavioral responses. The polymodal ASH sensory neurons are the primary nociceptors in C. elegans. We show here that the guanylyl cyclase ODR-1 functions non-cell-autonomously to downregulate ASH-mediated aversive behaviors and that ectopic cGMP generation in ASH is sufficient to dampen ASH sensitivity. We define a gap junction neural network that regulates nociception and propose that decentralized regulation of ASH signaling can allow for rapid correlation between an animal's internal state and its behavioral output, lending modulatory flexibility to this hard-wired nociceptive neural circuit.
Collapse
Affiliation(s)
- Michelle C. Krzyzanowski
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
| | - Sarah Woldemariam
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
| | - Jordan F. Wood
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
| | - Aditi H. Chaubey
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
| | - Chantal Brueggemann
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
| | - Alexander Bowitch
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
| | - Mary Bethke
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
| | - Noelle D. L’Etoile
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
| | - Denise M. Ferkey
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, New York, United States of America
| |
Collapse
|
28
|
Wehbi VL, Taskén K. Molecular Mechanisms for cAMP-Mediated Immunoregulation in T cells - Role of Anchored Protein Kinase A Signaling Units. Front Immunol 2016; 7:222. [PMID: 27375620 PMCID: PMC4896925 DOI: 10.3389/fimmu.2016.00222] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 05/23/2016] [Indexed: 12/20/2022] Open
Abstract
The cyclic AMP/protein kinase A (cAMP/PKA) pathway is one of the most common and versatile signal pathways in eukaryotic cells. A-kinase anchoring proteins (AKAPs) target PKA to specific substrates and distinct subcellular compartments providing spatial and temporal specificity for mediation of biological effects channeled through the cAMP/PKA pathway. In the immune system, cAMP is a potent negative regulator of T cell receptor-mediated activation of effector T cells (Teff) acting through a proximal PKA/Csk/Lck pathway anchored via a scaffold consisting of the AKAP Ezrin holding PKA, the linker protein EBP50, and the anchoring protein phosphoprotein associated with glycosphingolipid-enriched microdomains holding Csk. As PKA activates Csk and Csk inhibits Lck, this pathway in response to cAMP shuts down proximal T cell activation. This immunomodulating pathway in Teff mediates clinically important responses to regulatory T cell (Treg) suppression and inflammatory mediators, such as prostaglandins (PGs), adrenergic stimuli, adenosine, and a number of other ligands. A major inducer of T cell cAMP levels is PG E2 (PGE2) acting through EP2 and EP4 prostanoid receptors. PGE2 plays a crucial role in the normal physiological control of immune homeostasis as well as in inflammation and cancer immune evasion. Peripherally induced Tregs express cyclooxygenase-2, secrete PGE2, and elicit the immunosuppressive cAMP pathway in Teff as one tumor immune evasion mechanism. Moreover, a cAMP increase can also be induced by indirect mechanisms, such as intercellular transfer between T cells. Indeed, Treg, known to have elevated levels of intracellular cAMP, may mediate their suppressive function by transferring cAMP to Teff through gap junctions, which we speculate could also be regulated by PKA/AKAP complexes. In this review, we present an updated overview on the influence of cAMP-mediated immunoregulatory mechanisms acting through localized cAMP signaling and the therapeutical increasing prospects of AKAPs disruptors in T-cell immune function.
Collapse
Affiliation(s)
- Vanessa L. Wehbi
- Nordic EMBL Partnership, Centre for Molecular Medicine Norway, Oslo University Hospital, University of Oslo, Oslo, Norway
- Jebsen Inflammation Research Centre, Oslo University Hospital, Oslo, Norway
- Biotechnology Centre, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Kjetil Taskén
- Nordic EMBL Partnership, Centre for Molecular Medicine Norway, Oslo University Hospital, University of Oslo, Oslo, Norway
- Jebsen Inflammation Research Centre, Oslo University Hospital, Oslo, Norway
- Biotechnology Centre, Oslo University Hospital, University of Oslo, Oslo, Norway
- Jebsen Centre for Cancer Immunotherapy, Oslo University Hospital, Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
29
|
Schmidt K, Windler R, de Wit C. Communication Through Gap Junctions in the Endothelium. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 77:209-40. [PMID: 27451099 DOI: 10.1016/bs.apha.2016.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A swarm of fish displays a collective behavior (swarm behavior) and moves "en masse" despite the huge number of individual animals. In analogy, organ function is supported by a huge number of cells that act in an orchestrated fashion and this applies also to vascular cells along the vessel length. It is obvious that communication is required to achieve this vital goal. Gap junctions with their modular bricks, connexins (Cxs), provide channels that interlink the cytosol of adjacent cells by a pore sealed against the extracellular space. This allows the transfer of ions and charge and thereby the travel of membrane potential changes along the vascular wall. The endothelium provides a low-resistance pathway that depends crucially on connexin40 which is required for long-distance conduction of dilator signals in the microcirculation. The experimental evidence for membrane potential changes synchronizing vascular behavior is manifold but the functional verification of a physiologic role is still open. Other molecules may also be exchanged that possibly contribute to the synchronization (eg, Ca(2+)). Recent data suggest that vascular Cxs have more functions than just facilitating communication. As pharmacological tools to modulate gap junctions are lacking, Cx-deficient mice provide currently the standard to unravel their vascular functions. These include arteriolar dilation during functional hyperemia, hypoxic pulmonary vasoconstriction, vascular collateralization after ischemia, and feedback inhibition on renin secretion in the kidney.
Collapse
Affiliation(s)
- K Schmidt
- Institut für Physiologie, Universität zu Lübeck, Lübeck, Germany; Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V. (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - R Windler
- Institut für Physiologie, Universität zu Lübeck, Lübeck, Germany; Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V. (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - C de Wit
- Institut für Physiologie, Universität zu Lübeck, Lübeck, Germany; Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V. (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany.
| |
Collapse
|
30
|
Schadzek P, Schlingmann B, Schaarschmidt F, Lindner J, Koval M, Heisterkamp A, Preller M, Ngezahayo A. The cataract related mutation N188T in human connexin46 (hCx46) revealed a critical role for residue N188 in the docking process of gap junction channels. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1858:57-66. [PMID: 26449341 DOI: 10.1016/j.bbamem.2015.10.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 09/28/2015] [Accepted: 10/01/2015] [Indexed: 12/16/2022]
Abstract
The mutation N188T in human connexin46 (hCx46) correlates with a congenital nuclear pulverulent cataract. This mutation is in the second extracellular loop, a domain involved in docking of gap junction hemichannels. To analyze the functional consequences of this mutation, we expressed hCx46N188T and the wild type (hCx46wt) in Xenopus oocytes and HeLa cells. In Xenopus oocytes, hemichannels formed by hCx46wt and hCx46N188T had similar electrical properties. Additionally, a Ca(2+) and La(3+) sensitive current was observed in HeLa cells expressing eGFP-labeled hCx46wt or eGFP-labeled hCx46N188T. These results suggest that the N188T mutation did not alter apparent expression and the membrane targeting of the protein. Cells expressing hCx46wt-eGFP formed gap junction plaques, but plaques formed by hCx46N188T were extremely rare. A reduced plaque formation was also found in cells cotransfected with hCx46N188T-eGFP and mCherry-labeled hCx46wt as well as in cocultured cells expressing hCx46N188T-eGFP and hCx46wt-mCherry. Dye transfer experiments in cells expressing hCx46N188T revealed a lower transfer rate than cells expressing hCx46wt. We postulate that the N188T mutation affects intercellular connexon docking. This hypothesis is supported by molecular modeling of hCx46 using the crystal structure of hCx26 as a template. The model indicated that N188 is important for hemichannel docking through formation of hydrogen bonds with the residues R180, T189 and D191 of the opposing hCx46. The results suggest that the N188T mutation hinders the docking of the connexons to form gap junction channels. Moreover, the finding that a glutamine substitution (hCx46N188Q) could not rescue the docking emphasizes the specific role of N188.
Collapse
Affiliation(s)
- Patrik Schadzek
- Institute of Biophysics, Leibniz University Hannover, Germany
| | - Barbara Schlingmann
- Institute of Biophysics, Leibniz University Hannover, Germany; Division of Pulmonary, Allergy and Critical Care and Sleep Medicine, Department of Medicine and Department of Cell Biology, Emory School of Medicine, Atlanta, GA, USA
| | | | - Julia Lindner
- Institute of Biophysics, Leibniz University Hannover, Germany
| | - Michael Koval
- Division of Pulmonary, Allergy and Critical Care and Sleep Medicine, Department of Medicine and Department of Cell Biology, Emory School of Medicine, Atlanta, GA, USA; Department of Cell Biology, Emory University, Atlanta, GA, USA
| | | | - Matthias Preller
- Institute for Biophysical Chemistry, Hannover Medical School (MHH), Hannover, Germany; Center for Structural Systems Biology, German Electron Synchrotron (DESY), Hamburg, Germany.
| | - Anaclet Ngezahayo
- Institute of Biophysics, Leibniz University Hannover, Germany; Center for System Neurosciences (ZSN), Hannover, Germany.
| |
Collapse
|
31
|
Santa Cruz A, Meşe G, Valiuniene L, Brink PR, White TW, Valiunas V. Altered conductance and permeability of Cx40 mutations associated with atrial fibrillation. J Gen Physiol 2015; 146:387-98. [PMID: 26503720 PMCID: PMC4621748 DOI: 10.1085/jgp.201511475] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/23/2015] [Indexed: 11/20/2022] Open
Abstract
Gap junctions ensure the rapid propagation of the action potential throughout the myocardium. Three mutant forms of connexin40 (Cx40; A96S, M163V, and G38D), the primary component of the atrial gap junction channel, are associated with atrial fibrillation and retain the ability to form functional channels. We determined the biophysical properties of these mutant gap junctions in transiently transfected HeLa and N2A cells. All three mutants showed macroscopic junctional conductances over the range of 0.5 to 40 nS, and voltage dependences comparable to those of wild-type (WT) Cx40. However, the unitary conductance of G38D channels was ∼1.6-fold higher than that of WT Cx40 channels (∼220 vs. ∼135 pS), whereas the unitary conductances of the A96S and M163V mutants were similar to that of WT Cx40. Furthermore, the M163V and G38D channels exhibited approximately two- and approximately fivefold higher permeability to the anionic dye Lucifer yellow (LY) relative to K+ (LY/K+) compared with that of WT Cx40, whereas A96S LY transfer was similar to that of WT (G38D > M163V > A96S ≈ Cx40WT). In contrast, G38D channels were almost impermeable to cationic ethidium bromide (EtBr), suggesting that G38D alters channel selectivity. Conversely, A96S and M163V channels showed enhanced EtBr permeability relative to WT Cx40, with the following permeability order: M163V > A96S > Cx40WT > G38D. Altered conductive and permeability properties of mutant channels suggest an essential role for Cx40-mediated biochemical and electrical coupling in cardiac tissues. The altered properties of the three single-base substitution mutants may play a role in mechanisms of reentry arrhythmias.
Collapse
Affiliation(s)
- Ana Santa Cruz
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11792
| | - Gülistan Meşe
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11792
| | - Laima Valiuniene
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11792
| | - Peter R Brink
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11792
| | - Thomas W White
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11792
| | - Virginijus Valiunas
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11792
| |
Collapse
|
32
|
|
33
|
Abstract
Connexin mutations underlie numerous human genetic diseases. Several connexin genes have been linked to skin diseases, and mechanistic studies have indicated that a gain of abnormal channel function may be responsible for pathology. The topical accessibility of the epidermal connexins, the existence of several mouse models of human skin disease, and the ongoing identification of pharmacological inhibitors targeting connexins provide an opportunity to test new therapeutic approaches.
Collapse
Affiliation(s)
- Noah A Levit
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794, United States
| | - Thomas W White
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794, United States.
| |
Collapse
|
34
|
Connexins, gap junctions and peripheral neuropathy. Neurosci Lett 2015; 596:27-32. [DOI: 10.1016/j.neulet.2014.10.033] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/15/2014] [Accepted: 10/17/2014] [Indexed: 11/23/2022]
|
35
|
Bosen F, Schütz M, Beinhauer A, Strenzke N, Franz T, Willecke K. The Clouston syndrome mutation connexin30 A88V leads to hyperproliferation of sebaceous glands and hearing impairments in mice. FEBS Lett 2014; 588:1795-801. [DOI: 10.1016/j.febslet.2014.03.040] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 03/17/2014] [Accepted: 03/19/2014] [Indexed: 01/02/2023]
|
36
|
Billaud M, Lohman AW, Johnstone SR, Biwer LA, Mutchler S, Isakson BE. Regulation of cellular communication by signaling microdomains in the blood vessel wall. Pharmacol Rev 2014; 66:513-69. [PMID: 24671377 DOI: 10.1124/pr.112.007351] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
It has become increasingly clear that the accumulation of proteins in specific regions of the plasma membrane can facilitate cellular communication. These regions, termed signaling microdomains, are found throughout the blood vessel wall where cellular communication, both within and between cell types, must be tightly regulated to maintain proper vascular function. We will define a cellular signaling microdomain and apply this definition to the plethora of means by which cellular communication has been hypothesized to occur in the blood vessel wall. To that end, we make a case for three broad areas of cellular communication where signaling microdomains could play an important role: 1) paracrine release of free radicals and gaseous molecules such as nitric oxide and reactive oxygen species; 2) role of ion channels including gap junctions and potassium channels, especially those associated with the endothelium-derived hyperpolarization mediated signaling, and lastly, 3) mechanism of exocytosis that has considerable oversight by signaling microdomains, especially those associated with the release of von Willebrand factor. When summed, we believe that it is clear that the organization and regulation of signaling microdomains is an essential component to vessel wall function.
Collapse
Affiliation(s)
- Marie Billaud
- Dept. of Molecular Physiology and Biophysics, University of Virginia School of Medicine, PO Box 801394, Charlottesville, VA 22902.
| | | | | | | | | | | |
Collapse
|
37
|
Piccioni M, Chen Z, Tsun A, Li B. Regulatory T-cell differentiation and their function in immune regulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 841:67-97. [PMID: 25261205 DOI: 10.1007/978-94-017-9487-9_4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Regulatory T-cells (Treg) represent a subset of CD4+ T-cells characterized by high suppressive capacity, which can be generated in the thymus or induced in the periphery. The deleterious phenotype of the Scurfy mouse, which develops an X-linked lymphoproliferative disease resulting from defective T-cell tolerance, clearly demonstrates the importance of Treg cells for the maintenance of immune homeostasis. Although significant progress has been achieved, much information regarding the development, characteristics and function of Treg cells remain lacking. This chapter highlights the most recent discoveries in the field of Treg biology, focusing on the development and role of this cell subset in the maintenance of immune balance.
Collapse
Affiliation(s)
- Miranda Piccioni
- Key Laboratory of Molecular Virology and Immunology, Unit of Molecular Immunology, Institute Pasteur of Shanghai, Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
| | | | | | | |
Collapse
|
38
|
Chandrasekhar A, Kalmykov EA, Polusani SR, Mathis SA, Zucker SN, Nicholson BJ. Intercellular redistribution of cAMP underlies selective suppression of cancer cell growth by connexin26. PLoS One 2013; 8:e82335. [PMID: 24312655 PMCID: PMC3849486 DOI: 10.1371/journal.pone.0082335] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 10/30/2013] [Indexed: 12/02/2022] Open
Abstract
Connexins (Cx), which constitute gap junction intercellular channels in vertebrates, have been shown to suppress transformed cell growth and tumorigenesis, but the mechanism(s) still remain largely speculative. Here, we define the molecular basis by which Cx26, but less frequently Cx43 or Cx32, selectively confer growth suppression on cancer cells. Functional intercellular coupling is shown to be required, producing partial blocks of the cell cycle due to prolonged activation of several mitogenic kinases. PKA is both necessary and sufficient for the Cx26 induced growth inhibition in low serum and the absence of anchorage. Activation of PKA was not associated with elevated cAMP levels, but appeared to result from a redistribution of cAMP throughout the cell population, eliminating the cell cycle oscillations in cAMP required for efficient cell cycle progression. Cx43 and Cx32 fail to mediate this redistribution as, unlike Cx26, these channels are closed during the G2/M phase of the cell cycle when cAMP levels peak. Comparisons of tumor cell lines indicate that this is a general pattern, with growth suppression by connexins occurring whenever cAMP oscillates with the cell cycle, and the gap junction remain open throughout the cell cycle. Thus, gap junctional coupling, in the absence of any external signals, provides a general means to limit the mitotic rate of cell populations.
Collapse
Affiliation(s)
- Anjana Chandrasekhar
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Edward A. Kalmykov
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Srikanth R. Polusani
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Sandra A. Mathis
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Shoshanna N. Zucker
- Department of Pharmaceutical, Social and Administrative Sciences, D'Youville College School of Pharmacy,Buffalo, New York, United States of America
| | - Bruce J. Nicholson
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| |
Collapse
|
39
|
Haq N, Grose D, Ward E, Chiu O, Tigue N, Dowell SJ, Powell AJ, Chen MX. A high-throughput assay for connexin 43 (Cx43, GJA1) gap junctions using codon-optimized aequorin. Assay Drug Dev Technol 2012; 11:93-100. [PMID: 23046406 DOI: 10.1089/adt.2012.469] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Gap junctions (GJs) are intercellular channels which are composed of the connexin family of proteins that allow electrical and chemical communications and synchronization in tissue ensembles. Evidence suggests that pharmaceutical modulators of these channels may have therapeutic potential or carry undesired liability. In this report, we exogenously expressed human connexin 43 (Cx43, GJA1) and demonstrated functionality in a 96-well flow cytometry assay detecting intercellular transfer of the calcein dye. We have designed a 384-well high-throughput method for detecting the transfer of calcium between HeLa cells expressing Cx43. In this assay, donor cells coexpress Cx43 and the α1A adrenergic Gα-coupled receptor, while recipient cells coexpress Cx43 and the cytoplasmic version of the calcium-sensitive luminescent protein aequorin enhanced by codon optimization (cytoAeq). The two cell populations were mixed, dispensed to 384-well plates, and incubated for 3 h to allow the formation of GJs. Activation of α1A by epinephrine in donor cells led to dose-dependent calcium increases in recipient cells, which were detected by measuring the intensity of aequorin luminescence. The response was dependent on the expression of Cx43 and inhibited by the GJ blocker 18α-glycyrrhetinic acid, suggesting Cx43 GJ-mediated activity. In a parallel experiment with capsaicin and the TrpV1 ion channel in place of phenylephrine and α1A, a similar magnitude of difference in the maximal calcium response was detected in both donor and recipient cells, suggesting that calcium is likely the permeant ion through the GJ. This assay may pave the way for high-throughput screening of GJ modulators for drug discovery.
Collapse
Affiliation(s)
- Nazia Haq
- Biological Reagents and Assay Development, GlaxoSmithKline R&D, Stevenage, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
40
|
KERR PAULM, TAM RAYMOND, ONDRUSOVA KATARINA, MITTAL ROHAN, NARANG DEEPAK, TRAN CAMHAT, WELSH DONALDG, PLANE FRANCES. Endothelial Feedback and the Myoendothelial Projection. Microcirculation 2012; 19:416-22. [DOI: 10.1111/j.1549-8719.2012.00187.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
41
|
Bukauskas FF. Neurons and β-cells of the pancreas express connexin36, forming gap junction channels that exhibit strong cationic selectivity. J Membr Biol 2012; 245:243-53. [PMID: 22752717 PMCID: PMC3626077 DOI: 10.1007/s00232-012-9445-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 06/01/2012] [Indexed: 01/30/2023]
Abstract
We examined the permeability of connexin36 (Cx36) homotypic gap junction (GJ) channels, expressed in neurons and β-cells of the pancreas, to dyes differing in molecular mass and net charge. Experiments were performed in HeLa cells stably expressing Cx36 tagged with EGFP by combining a dual whole-cell voltage clamp and fluorescence imaging. To assess the permeability of the single GJ channel (P(γ)), we used a dual-mode excitation of fluorescent dyes that allowed us to measure cell-to-cell dye transfer at levels not resolvable using whole-field excitation solely. We demonstrate that P(γ) of Cx36 for cationic dyes (EAM-1⁺ and EAM-2⁺) is ~10-fold higher than that for an anionic dye of the same net charge and similar molecular mass, Alexa fluor-350 (AFl-350⁻). In addition, P(γ) for Lucifer yellow (LY²⁻) is approximately fourfold smaller than that for AFl-350⁻, which suggests that the higher negativity of LY²⁻ significantly reduces permeability. The P(γ) of Cx36 for AFl-350 is approximately 358, 138, 23 and four times smaller than the P(γ)s of Cx43, Cx40, Cx45, and Cx57, respectively. In contrast, it is 6.5-fold higher than the P(γ) of mCx30.2, which exhibits a smaller single-channel conductance. Thus, Cx36 GJs are highly cation-selective and should exhibit relatively low permeability to numerous vital negatively charged metabolites and high permeability to K⁺, a major charge carrier in cell-cell communication.
Collapse
Affiliation(s)
- Feliksas F Bukauskas
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| |
Collapse
|
42
|
Degen J, Schütz M, Dicke N, Strenzke N, Jokwitz M, Moser T, Willecke K. Connexin32 can restore hearing in connexin26 deficient mice. Eur J Cell Biol 2011; 90:817-24. [PMID: 21813206 DOI: 10.1016/j.ejcb.2011.05.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 04/20/2011] [Accepted: 05/03/2011] [Indexed: 12/15/2022] Open
Abstract
Functional gap junction channels composed of certain connexin proteins are essential for the function of the cochlea. Homozygous deficiency in the Gjb2 (mice) or GJB2 (human) gene coding for connexin26 (Cx26) in the cochlea leads to hearing impairment in mice and humans, respectively. Here we have studied the functional equivalence of Cx26 and connexin32 (Cx32) isoforms in the cochlea. We analyzed a conditional mouse mutant in which the Gjb2 coding DNA was exchanged by LacZ DNA coding for the reporter protein beta-galactosidase. This allowed us to follow the unrestricted and cell type specific expression of Gjb2 promoter activity. After inner ear specific, Otogelin-Cre recombinase mediated deletion of the loxP-site-flanked LacZ coding DNA, transcription of the Gjb1 gene, coding for Cx32 was activated by the Gjb2 promoter. Interbreeding of these mice with conditional Gjb2 null mice resulted in animals in which Cx32 instead of Cx26 protein is expressed in the non-sensory epithelial network of the cochlea. When we analyzed the auditory function in these mice, we found that the expression of Cx32 protein is sufficient to support hearing in the absence of Cx26. Thus Cx32 can functionally replace Cx26 in the mouse cochlea resulting in almost normal hearing.
Collapse
Affiliation(s)
- Joachim Degen
- Institute of Genetics, University of Bonn, 53117 Bonn, Germany
| | | | | | | | | | | | | |
Collapse
|
43
|
Shaikh MA, Wall DJN, David T. Macro-scale phenomena of arterial coupled cells: a massively parallel simulation. J R Soc Interface 2011; 9:972-87. [PMID: 21920960 DOI: 10.1098/rsif.2011.0453] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Impaired mass transfer characteristics of blood-borne vasoactive species such as adenosine triphosphate in regions such as an arterial bifurcation have been hypothesized as a prospective mechanism in the aetiology of atherosclerotic lesions. Arterial endothelial cells (ECs) and smooth muscle cells (SMCs) respond differentially to altered local haemodynamics and produce coordinated macro-scale responses via intercellular communication. Using a computationally designed arterial segment comprising large populations of mathematically modelled coupled ECs and SMCs, we investigate their response to spatial gradients of blood-borne agonist concentrations and the effect of micro-scale-driven perturbation on the macro-scale. Altering homocellular (between same cell type) and heterocellular (between different cell types) intercellular coupling, we simulated four cases of normal and pathological arterial segments experiencing an identical gradient in the concentration of the agonist. Results show that the heterocellular calcium (Ca(2+)) coupling between ECs and SMCs is important in eliciting a rapid response when the vessel segment is stimulated by the agonist gradient. In the absence of heterocellular coupling, homocellular Ca(2+) coupling between SMCs is necessary for propagation of Ca(2+) waves from downstream to upstream cells axially. Desynchronized intracellular Ca(2+) oscillations in coupled SMCs are mandatory for this propagation. Upon decoupling the heterocellular membrane potential, the arterial segment looses the inhibitory effect of ECs on the Ca(2+) dynamics of the underlying SMCs. The full system comprises hundreds of thousands of coupled nonlinear ordinary differential equations simulated on the massively parallel Blue Gene architecture. The use of massively parallel computational architectures shows the capability of this approach to address macro-scale phenomena driven by elementary micro-scale components of the system.
Collapse
Affiliation(s)
- Mohsin Ahmed Shaikh
- Center for Bioengineering, University of Canterbury, Christchurch, New Zealand.
| | | | | |
Collapse
|
44
|
Palacios-Prado N, Bukauskas FF. Modulation of metabolic communication through gap junction channels by transjunctional voltage; synergistic and antagonistic effects of gating and ionophoresis. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2011; 1818:1884-94. [PMID: 21930112 DOI: 10.1016/j.bbamem.2011.09.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Revised: 08/25/2011] [Accepted: 09/02/2011] [Indexed: 01/28/2023]
Abstract
Gap junction (GJ) channels assembled from connexin (Cx) proteins provide a structural basis for direct electrical and metabolic cell-cell communication. Here, we focus on gating and permeability properties of Cx43/Cx45 heterotypic GJs exhibiting asymmetries of both voltage-gating and transjunctional flux (J(j)) of fluorescent dyes depending on transjunctional voltage (V(j)). Relatively small differences in the resting potential of communicating cells can substantially reduce or enhance this flux at relative negativity or positivity on Cx45 side, respectively. Similarly, series of V(j) pulses resembling bursts of action potentials (APs) reduce J(j) when APs initiate in the cell expressing Cx43 and increase J(j) when APs initiate in the cell expressing Cx45. J(j) of charged fluorescent dyes is affected by ionophoresis and V(j)-gating and the asymmetry of J(j)-V(j) dependence in heterotypic GJs is enhanced or reduced when ionophoresis and V(j)-gating work in a synergistic or antagonistic manner, respectively. Modulation of cell-to-cell transfer of metabolites and signaling molecules by V(j) may occur in excitable as well as non-excitable tissues and may be more expressed in the border between normal and pathological regions where intercellular gradients of membrane potential and concentration of ions are substantially altered. This article is part of a Special Issue entitled: The Communicating junctions, composition, structure and characteristics.
Collapse
Affiliation(s)
- Nicolás Palacios-Prado
- Dominick P.Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | | |
Collapse
|
45
|
Abstract
The aim of the present study was to show the participation and physiological role of calmodulin (CaM) and cAMP during vitellogenin endocytic uptake in the amphibian Xenopus laevis. The results showed a differential distribution of CaM in the ovary follicles during oogenesis. The CaM intracellular localization was not affected by gap junction's downregulation and CaM inhibition did not completely abolished the endocytic activity of oocytes. We showed that cAMP was able to completely rescue the endocytic competence in follicles in which gap junctional communication had been disrupted by octanol. Moreover cAMP was capable of restoring oocyte endocytic capability in the presence of octanol and stelazine, a CaM inhibitor. We propose that, in Vtg uptake regulation, cAMP is upstream of CaM during the endocytic signalling pathway.
Collapse
|
46
|
Maeda S, Tsukihara T. Structure of the gap junction channel and its implications for its biological functions. Cell Mol Life Sci 2011; 68:1115-29. [PMID: 20960023 PMCID: PMC11114897 DOI: 10.1007/s00018-010-0551-z] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Revised: 09/28/2010] [Accepted: 09/30/2010] [Indexed: 12/16/2022]
Abstract
Gap junctions consist of arrays of intercellular channels composed of integral membrane proteins called connexin in vertebrates. Gap junction channels regulate the passage of ions and biological molecules between adjacent cells and, therefore, are critically important in many biological activities, including development, differentiation, neural activity, and immune response. Mutations in connexin genes are associated with several human diseases, such as neurodegenerative disease, skin disease, deafness, and developmental abnormalities. The activity of gap junction channels is regulated by the membrane voltage, intracellular microenvironment, interaction with other proteins, and phosphorylation. Each connexin channel has its own property for conductance and molecular permeability. A number of studies have tried to reveal the molecular architecture of the channel pore that should confer the connexin-specific permeability/selectivity properties and molecular basis for the gating and regulation. In this review, we give an overview of structural studies and describe the structural and functional relationship of gap junction channels.
Collapse
Affiliation(s)
- Shoji Maeda
- Institute for Protein Research, Osaka University, OLABB, 6-2-3 Furuedai, Suita, 565-0874 Japan
- Department of Life Science, University of Hyogo, 3-2-1 Koto, Kamighori, Akoh, Hyogo 678-1297 Japan
- Present Address: Paul Scherrer Institut, Biology and Chemistry OFLG 101, 5232 Villigen, Switzerland
| | - Tomitake Tsukihara
- Institute for Protein Research, Osaka University, OLABB, 6-2-3 Furuedai, Suita, 565-0874 Japan
- Department of Life Science, University of Hyogo, 3-2-1 Koto, Kamighori, Akoh, Hyogo 678-1297 Japan
| |
Collapse
|
47
|
von Maltzahn J, Wulf V, Matern G, Willecke K. Connexin39 deficient mice display accelerated myogenesis and regeneration of skeletal muscle. Exp Cell Res 2011; 317:1169-78. [PMID: 21272575 DOI: 10.1016/j.yexcr.2011.01.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 01/14/2011] [Accepted: 01/15/2011] [Indexed: 12/19/2022]
Abstract
During muscle development and regeneration of skeletal muscle in mice connexin43 (Cx43) and connexin39 (Cx39) are specifically expressed: Cx43 in satellite cells and myoblasts, whereas Cx39 is exclusively expressed in myogenin-positive cells. We generated Cx39 deficient mice by replacing the coding region of the Gjd4 gene by DNA coding for the enhanced green fluorescent protein eGFP. Adult Cx39 deficient mice exhibit no obvious phenotypic alterations of skeletal muscle compared to wild type mice in the resting state. However, myogenesis in Cx39 deficient embryos is accelerated as indicated by increased myogenin expression on ED13.5 and ED16.5 and increased expression of Cx43 in developing skeletal muscle. In addition, the regeneration process of skeletal muscle in Cx39 deficient mice is accelerated as shown by a 2day earlier onset of MyoD and myogenin expression, relative to wild type littermates. Interestingly, Cx43 expression was also upregulated in Cx39 deficient mice during regeneration of skeletal muscle. We hypothesize that Cx43 may compensate for the loss of Cx39 during myogenesis and regeneration.
Collapse
|
48
|
Kanaporis G, Brink PR, Valiunas V. Gap junction permeability: selectivity for anionic and cationic probes. Am J Physiol Cell Physiol 2010; 300:C600-9. [PMID: 21148413 DOI: 10.1152/ajpcell.00316.2010] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Gap junction channels formed by different connexins exhibit specific permeability to a variety of larger solutes including second messengers, polypeptides, and small interfering RNAs. Here, we report the permeability of homotypic connexin26 (Cx26), Cx40, Cx43, and Cx45 gap junction channels stably expressed in HeLa cells to solutes with different size and net charge. Channel permeability was determined using simultaneous measurements of junctional conductance and the cell-cell flux of a fluorescent probe. All four connexins allowed passage of both cationic and anionic probes, but the transfer rates were connexin dependent. The negatively charged probes [Lucifer yellow (LY; median axial diameter 9.9 Å, charge -2), carboxyfluorescein (CF; 8.2 Å; -2), and Alexa Fluor350 (AF350, 5.4 Å; -1)] exhibited the following permeability order: Cx43 > Cx45 > Cx26 > Cx40. In contrast, for the positively charged species permeability, the orders were as follows: Cx26 ≈ Cx43 ≈ Cx40 ≈ Cx45 for N,N,N-trimethyl-2-[methyl-(7-nitro-2,1,3-benzoxadiol-4-yl) amino] ethanaminium (NBD-m-TMA; 5.5 Å, +1) and Cx26 ≥ Cx43 ≈ Cx40 > Cx45 for ethidium bromide (10.3 Å, +1). Comparison of probe permeability relative to K(+) revealed that Cx43 and Cx45 exhibited similar permeability for NBD-m-TMA and AF350, indicating weak charge selectivity. However, lesser transfer of CF and LY through Cx45 relative to Cx43 channels suggests stronger size-dependent discrimination of solute. The permeability of NBD-m-TMA for Cx40 and Cx26 channels was approximately three times higher than to anionic AF350 despite the fact that both have similar minor diameters, suggesting charge selectivity. In conclusion, these results confirm that channels formed from individual connexins can discriminate for solutes based on size and charge, suggesting that channel selectivity may be a key factor in cell signaling.
Collapse
Affiliation(s)
- G Kanaporis
- Dept. of Physiology and Biophysics, State University of New York at Stony Brook, Stony Brook, NY 11794-8661, USA
| | | | | |
Collapse
|
49
|
Frank M, Eiberger B, Janssen-Bienhold U, de Sevilla Müller LP, Tjarks A, Kim JS, Maschke S, Dobrowolski R, Sasse P, Weiler R, Fleischmann BK, Willecke K. Neuronal connexin-36 can functionally replace connexin-45 in mouse retina but not in the developing heart. J Cell Sci 2010; 123:3605-15. [PMID: 20930146 DOI: 10.1242/jcs.068668] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023] Open
Abstract
The gap junction protein connexin-45 (Cx45) is expressed in the conduction system of the heart and in certain neurons of the retina and brain. General and cardiomyocyte-directed deficiencies of Cx45 in mice lead to lethality on embryonic day 10.5 as a result of cardiovascular defects. Neuron-directed deletion of Cx45 leads to defects in transmission of visual signals. Connexin-36 (Cx36) is co-expressed with Cx45 in certain types of retinal interneurons. To determine whether these two connexins have similar functions and whether Cx36 can compensate for Cx45, we generated knock-in mice in which DNA encoding Cx45 was replaced with that encoding Cx36. Neuron-directed replacement of Cx45 with Cx36 resulted in viable animals. Electroretinographic and neurotransmitter coupling analyses demonstrated functional compensation in the retina. By contrast, general and cardiomyocyte-directed gene replacement led to lethality on embryonic day 11.5. Mutant embryos displayed defects in cardiac morphogenesis and conduction. Thus, functional compensation of Cx45 by Cx36 did not occur during embryonic heart development. These data suggest that Cx45 and Cx36 have similar functions in the retina, whereas Cx45 fulfills special functions in the developing heart that cannot be compensated by Cx36.
Collapse
Affiliation(s)
- Marina Frank
- Institute of Genetics, University of Bonn, 53117 Bonn, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Bopp T, Radsak M, Schmitt E, Schild H. New strategies for the manipulation of adaptive immune responses. Cancer Immunol Immunother 2010; 59:1443-8. [PMID: 20361184 PMCID: PMC11030961 DOI: 10.1007/s00262-010-0851-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Accepted: 03/18/2010] [Indexed: 12/19/2022]
Abstract
The maintenance of peripheral tolerance is largely based on thymus-derived CD4(+)CD25(+) naturally occurring regulatory T cells (Tregs). While on the one hand being indispensable for the perpetuation of tolerance to self-antigens, the immune suppressive properties of Tregs contribute to cancer pathogenesis and progression. Thus, modulation of Treg function represents a promising strategy to support tumor eradication in immunotherapy of cancer. Here, we discuss potential therapeutic applications of our observation that Tregs contain high concentrations of the second messenger cyclic adenosine monophosphate, which is transferred from Tregs via gap junctions to suppress the function of T cells and dendritic cells.
Collapse
Affiliation(s)
- Tobias Bopp
- Institute for Immunology, University Medical Centre of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Markus Radsak
- Institute for Immunology, University Medical Centre of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- III-Medical Clinic, University Medical Centre of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Edgar Schmitt
- Institute for Immunology, University Medical Centre of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Hansjörg Schild
- Institute for Immunology, University Medical Centre of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| |
Collapse
|