51
|
Vallon V, Unwin R, Inscho EW, Leipziger J, Kishore BK. Extracellular Nucleotides and P2 Receptors in Renal Function. Physiol Rev 2019; 100:211-269. [PMID: 31437091 DOI: 10.1152/physrev.00038.2018] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The understanding of the nucleotide/P2 receptor system in the regulation of renal hemodynamics and transport function has grown exponentially over the last 20 yr. This review attempts to integrate the available data while also identifying areas of missing information. First, the determinants of nucleotide concentrations in the interstitial and tubular fluids of the kidney are described, including mechanisms of cellular release of nucleotides and their extracellular breakdown. Then the renal cell membrane expression of P2X and P2Y receptors is discussed in the context of their effects on renal vascular and tubular functions. Attention is paid to effects on the cortical vasculature and intraglomerular structures, autoregulation of renal blood flow, tubuloglomerular feedback, and the control of medullary blood flow. The role of the nucleotide/P2 receptor system in the autocrine/paracrine regulation of sodium and fluid transport in the tubular and collecting duct system is outlined together with its role in integrative sodium and fluid homeostasis and blood pressure control. The final section summarizes the rapidly growing evidence indicating a prominent role of the extracellular nucleotide/P2 receptor system in the pathophysiology of the kidney and aims to identify potential therapeutic opportunities, including hypertension, lithium-induced nephropathy, polycystic kidney disease, and kidney inflammation. We are only beginning to unravel the distinct physiological and pathophysiological influences of the extracellular nucleotide/P2 receptor system and the associated therapeutic perspectives.
Collapse
Affiliation(s)
- Volker Vallon
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Robert Unwin
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Edward W Inscho
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Jens Leipziger
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Bellamkonda K Kishore
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| |
Collapse
|
52
|
Stožer A, Markovič R, Dolenšek J, Perc M, Marhl M, Slak Rupnik M, Gosak M. Heterogeneity and Delayed Activation as Hallmarks of Self-Organization and Criticality in Excitable Tissue. Front Physiol 2019; 10:869. [PMID: 31333504 PMCID: PMC6624746 DOI: 10.3389/fphys.2019.00869] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 06/21/2019] [Indexed: 12/14/2022] Open
Abstract
Self-organized critical dynamics is assumed to be an attractive mode of functioning for several real-life systems and entails an emergent activity in which the extent of observables follows a power-law distribution. The hallmarks of criticality have recently been observed in a plethora of biological systems, including beta cell populations within pancreatic islets of Langerhans. In the present study, we systematically explored the mechanisms that drive the critical and supercritical behavior in networks of coupled beta cells under different circumstances by means of experimental and computational approaches. Experimentally, we employed high-speed functional multicellular calcium imaging of fluorescently labeled acute mouse pancreas tissue slices to record calcium signals in a large number of beta cells simultaneously, and with a high spatiotemporal resolution. Our experimental results revealed that the cellular responses to stimulation with glucose are biphasic and glucose-dependent. Under physiological as well as under supraphysiological levels of stimulation, an initial activation phase was followed by a supercritical plateau phase with a high number of global intercellular calcium waves. However, the activation phase displayed fingerprints of critical behavior under lower stimulation levels, with a progressive recruitment of cells and a power-law distribution of calcium wave sizes. On the other hand, the activation phase provoked by pathophysiologically high glucose concentrations, differed considerably and was more rapid, less continuous, and supercritical. To gain a deeper insight into the experimentally observed complex dynamical patterns, we built up a phenomenological model of coupled excitable cells and explored empirically the model’s necessities that ensured a good overlap between computational and experimental results. It turned out that such a good agreement between experimental and computational findings was attained when both heterogeneous and stimulus-dependent time lags, variability in excitability levels, as well as a heterogeneous cell-cell coupling were included into the model. Most importantly, since our phenomenological approach involved only a few parameters, it naturally lends itself not only for determining key mechanisms of self-organized criticality at the tissue level, but also points out various features for comprehensive and realistic modeling of different excitable systems in nature.
Collapse
Affiliation(s)
- Andraž Stožer
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Rene Markovič
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia.,Faculty of Education, University of Maribor, Maribor, Slovenia.,Faculty of Energy Technology, University of Maribor, Krško, Slovenia
| | - Jurij Dolenšek
- Faculty of Medicine, University of Maribor, Maribor, Slovenia.,Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | - Matjaž Perc
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia.,Center for Applied Mathematics and Theoretical Physics, University of Maribor, Maribor, Slovenia.,Complexity Science Hub Vienna, Vienna, Austria
| | - Marko Marhl
- Faculty of Medicine, University of Maribor, Maribor, Slovenia.,Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia.,Faculty of Education, University of Maribor, Maribor, Slovenia
| | - Marjan Slak Rupnik
- Faculty of Medicine, University of Maribor, Maribor, Slovenia.,Institute of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.,Alma Mater Europaea - ECM, Maribor, Slovenia
| | - Marko Gosak
- Faculty of Medicine, University of Maribor, Maribor, Slovenia.,Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| |
Collapse
|
53
|
Le Gal L, Pellegrin M, Santoro T, Mazzolai L, Kurtz A, Meda P, Wagner C, Haefliger J. Connexin37-Dependent Mechanisms Selectively Contribute to Modulate Angiotensin II -Mediated Hypertension. J Am Heart Assoc 2019; 8:e010823. [PMID: 30943815 PMCID: PMC6507190 DOI: 10.1161/jaha.118.010823] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/30/2019] [Indexed: 12/23/2022]
Abstract
Background Gap junction channels made of Connexin37 (Cx37) are expressed by aortic endothelial and smooth muscle cells of hypertensive mice, as well as by the renin-secreting cells of kidneys. Methods and Results To decipher whether Cx37 has any role in hypertension, angiotensin II (Ang II ) was infused in normotensive wild-type and Cx37-deficient mice (Cx37-/-). After 2 to 4 weeks, the resulting increase in blood pressure was lower in Cx37-/- than in wild-type mice, suggesting an alteration in the Ang II response. To investigate this possibility, mice were submitted to a 2-kidney, 1-clip procedure, a renin-dependent model of hypertension. Two weeks after this clipping, Cx37-/- mice were less hypertensive than wild-type mice and, 2 weeks later, their blood pressure had returned to control values, in spite of abnormally high plasma renin levels. In contrast, Cx37-/- and wild-type mice that received N-nitro-l-arginine-methyl-ester, a renin-independent model of hypertension, featured a similar and sustained increase in blood pressure. The data indicate that loss of Cx37 selectively altered the Ang II -dependent pathways. Consistent with this conclusion, aortas of Cx37-/- mice featured an increased basal expression of the Ang II type 2 receptors ( AT 2R), and increased transcripts levels of downstream signaling proteins, such as Cnksr1 and Ptpn6 ( SHP -1). Accordingly, the response of Cx37-/- mice aortas to an ex vivo Ang II exposure was altered, since phosphorylation levels of several proteins of the Ang II pathway ( MLC 2, ERK , and AKT ) remained unchanged. Conclusions These findings provide evidence that Cx37 selectively influences Ang II signaling, mostly via a modulation of the expression of the Ang II type 2 receptor.
Collapse
Affiliation(s)
- Loïc Le Gal
- Department of MedicineUniversity of LausanneSwitzerland
| | - Maxime Pellegrin
- Division of AngiologyHeart and Vessel DepartmentCentre Hospitalier Universitaire VaudoisUniversity of LausanneSwitzerland
| | - Tania Santoro
- Department of MedicineUniversity of LausanneSwitzerland
| | - Lucia Mazzolai
- Division of AngiologyHeart and Vessel DepartmentCentre Hospitalier Universitaire VaudoisUniversity of LausanneSwitzerland
| | - Armin Kurtz
- Department of PhysiologyUniversity of RegensburgGermany
| | - Paolo Meda
- Department of Cell Physiology and MetabolismSchool of MedicineCMUUniversity of GenevaSwitzerland
| | | | | |
Collapse
|
54
|
Ranjbaran A, Latifi Z, Nejabati HR, Abroon S, Mihanfar A, Sadigh AR, Fattahi A, Nouri M, Raffel N. Exosome‐based intercellular communication in female reproductive microenvironments. J Cell Physiol 2019; 234:19212-19222. [DOI: 10.1002/jcp.28668] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 03/25/2019] [Accepted: 04/02/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Ali Ranjbaran
- Women's Reproductive Health Research Center Tabriz University of Medical Sciences Tabriz Iran
| | - Zeinab Latifi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medical Sciences Tabriz University of Medical Sciences Tabriz Iran
| | - Hamid Reza Nejabati
- Department of Biochemistry and Clinical Laboratories, Faculty of Medical Sciences Tabriz University of Medical Sciences Tabriz Iran
| | - Sina Abroon
- Stem Cell Research Center Tabriz University of Medical Sciences Tabriz Iran
| | - Aynaz Mihanfar
- Department of Biochemistry, Faculty of Medicine Urmia University of Medical Sciences Urmia Iran
| | - Aydin Raei Sadigh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medical Sciences Tabriz University of Medical Sciences Tabriz Iran
| | - Amir Fattahi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences Tabriz University of Medical Sciences Tabriz Iran
- Student Research Committee Tabriz University of Medical Sciences Tabriz Iran
| | - Mohammad Nouri
- Stem Cell Research Center Tabriz University of Medical Sciences Tabriz Iran
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences Tabriz University of Medical Sciences Tabriz Iran
| | - Nathalie Raffel
- Department of Obstetrics and Gynecology Erlangen University Hospital, Friedrich‐Alexander‐Universität Erlangen‐Nürnberg Erlangen Germany
| |
Collapse
|
55
|
Khalyfa A, Gozal D. Connexins and Atrial Fibrillation in Obstructive Sleep Apnea. CURRENT SLEEP MEDICINE REPORTS 2018; 4:300-311. [PMID: 31106116 PMCID: PMC6516763 DOI: 10.1007/s40675-018-0130-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF THE REVIEW To summarize the potential interactions between obstructive sleep apnea (OSA), atrial fibrillation (AF), and connexins. RECENT FINDINGS OSA is highly prevalent in patients with cardiovascular disease, and is associated with increased risk for end-organ substantial morbidities linked to autonomic nervous system imbalance, increased oxidative stress and inflammation, ultimately leading to reduced life expectancy. Epidemiological studies indicate that OSA is associated with increased incidence and progression of coronary heart disease, heart failure, stroke, as well as arrhythmias, particularly AF. Conversely, AF is very common among subjects referred for suspected OSA, and the prevalence of AF increases with OSA severity. The interrelationships between AF and OSA along with the well-known epidemiological links between these two conditions and obesity may reflect shared pathophysiological pathways, which may depend on the intercellular diffusion of signaling molecules into either the extracellular space or require cell-to-cell contact. Connexin signaling is accomplished via direct exchanges of cytosolic molecules between adjacent cells at gap membrane junctions for cell-to-cell coupling. The role of connexins in AF is now quite well established, but the impact of OSA on cardiac connexins has only recently begun to be investigated. Understanding the biology and regulatory mechanisms of connexins in OSA at the transcriptional, translational, and post-translational levels will undoubtedly require major efforts to decipher the breadth and complexity of connexin functions in OSA-induced AF. SUMMARY The risk of end-organ morbidities has initiated the search for circulating mechanistic biomarker signatures and the implementation of biomarker-based algorithms for precision-based diagnosis and risk assessment. Here we summarize recent findings in OSA as they relate to AF risk, and also review potential mechanisms linking OSA, AF and connexins.
Collapse
Affiliation(s)
- Abdelnaby Khalyfa
- Department of Pediatrics, Biological Sciences Division, Pritzker School of Medicine, The University of Chicago, Chicago IL 60637, USA
| | - David Gozal
- Department of Child Health, University of Missouri School of Medicine, Columbia, MO 65201, USA
| |
Collapse
|
56
|
Castorena-Gonzalez JA, Zawieja SD, Li M, Srinivasan RS, Simon AM, de Wit C, de la Torre R, Martinez-Lemus LA, Hennig GW, Davis MJ. Mechanisms of Connexin-Related Lymphedema. Circ Res 2018; 123:964-985. [PMID: 30355030 PMCID: PMC6771293 DOI: 10.1161/circresaha.117.312576] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
RATIONALE Mutations in GJC2 and GJA1, encoding Cxs (connexins) 47 and 43, respectively, are linked to lymphedema, but the underlying mechanisms are unknown. Because efficient lymph transport relies on the coordinated contractions of lymphatic muscle cells (LMCs) and their electrical coupling through Cxs, Cx-related lymphedema is proposed to result from dyssynchronous contractions of lymphatic vessels. OBJECTIVE To determine which Cx isoforms in LMCs and lymphatic endothelial cells are required for the entrainment of lymphatic contraction waves and efficient lymph transport. METHODS AND RESULTS We developed novel methods to quantify the spatiotemporal entrainment of lymphatic contraction waves and used optogenetic techniques to analyze calcium signaling within and between the LMC and the lymphatic endothelial cell layers. Genetic deletion of the major lymphatic endothelial cell Cxs (Cx43, Cx47, or Cx37) revealed that none were necessary for the synchronization of the global calcium events that triggered propagating contraction waves. We identified Cx45 in human and mouse LMCs as the critical Cx mediating the conduction of pacemaking signals and entrained contractions. Smooth muscle-specific Cx45 deficiency resulted in 10- to 18-fold reduction in conduction speed, partial-to-severe loss of contractile coordination, and impaired lymph pump function ex vivo and in vivo. Cx45 deficiency resulted in profound inhibition of lymph transport in vivo, but only under an imposed gravitational load. CONCLUSIONS Our results (1) identify Cx45 as the Cx isoform mediating the entrainment of the contraction waves in LMCs; (2) show that major endothelial Cxs are dispensable for the entrainment of contractions; (3) reveal a lack of coupling between lymphatic endothelial cells and LMCs, in contrast to arterioles; (4) point to lymphatic valve defects, rather than contraction dyssynchrony, as the mechanism underlying GJC2- or GJA1-related lymphedema; and (5) show that a gravitational load exacerbates lymphatic contractile defects in the intact mouse hindlimb, which is likely critical for the development of lymphedema in the adult mouse.
Collapse
Affiliation(s)
| | - Scott D. Zawieja
- Dept. of Medical Pharmacology and Physiology and University of Missouri School of Medicine
| | - Min Li
- Dept. of Medical Pharmacology and Physiology and University of Missouri School of Medicine
| | - R. Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City OK
| | | | - Cor de Wit
- Institute of Physiology, University of Luebeck, Luebeck Germany
| | | | - Luis A. Martinez-Lemus
- Dept. of Medical Pharmacology and Physiology and University of Missouri School of Medicine
| | | | - Michael J. Davis
- Dept. of Medical Pharmacology and Physiology and University of Missouri School of Medicine
| |
Collapse
|
57
|
Oshima A. Structure of an innexin gap junction channel and cryo-EM sample preparation. Microscopy (Oxf) 2018; 66:371-379. [PMID: 29036409 PMCID: PMC6084585 DOI: 10.1093/jmicro/dfx035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 08/31/2017] [Indexed: 01/05/2023] Open
Abstract
Gap junction channels are essential for mediating intercellular communication in most multicellular organisms. Two gene families encode gap junction channels, innexin and connexin. Although the sequence similarity between these two families based on bioinformatics is not conclusively determined, the gap junction channels encoded by these two gene families are structurally and functionally analogous. We recently reported an atomic structure of an invertebrate innexin gap junction channel using single-particle cryo-electron microscopy. Our findings revealed that connexin and innexin families share several structural properties with regard to their monomeric and oligomeric structures, while simultaneously suggesting a diversity of gap junction channels in nature. This review summarizes cutting-edge progress toward determining an innexin gap junction channel structure, as well as essential tips for preparing cryo-electron microscopy samples for high-resolution structural analysis of an innexin gap junction channel.
Collapse
Affiliation(s)
- Atsunori Oshima
- Cellular and Structural Physiology Institute (CeSPI), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan.,Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| |
Collapse
|
58
|
Ray A, Katoch P, Jain N, Mehta PP. Dileucine-like motifs in the C-terminal tail of connexin32 control its endocytosis and assembly into gap junctions. J Cell Sci 2018; 131:jcs207340. [PMID: 29361528 PMCID: PMC5897717 DOI: 10.1242/jcs.207340] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 01/11/2018] [Indexed: 12/11/2022] Open
Abstract
Defects in assembly of gap junction-forming proteins, called connexins (Cxs), are observed in a variety of cancers. Connexin32 (Cx32; also known as GJB1) is expressed by the polarized cells in epithelia. We discovered two dileucine-based motifs, which govern the intracellular sorting and endocytosis of transmembrane proteins, in the C-terminal tail of Cx32 and explored their role in regulating its endocytosis and gap junction-forming abilities in pancreatic and prostate cancer cells. One motif, designated as LI, was located near the juxtamembrane domain, whereas the other, designated as LL, was located distally. We also discovered a non-canonical motif, designated as LR, in the C-terminal tail. Our results showed that rendering these motifs non-functional had no effect on the intracellular sorting of Cx32. However, rendering the LL or LR motif nonfunctional enhanced the formation of gap junctions by inhibiting Cx32 endocytosis by the clathrin-mediated pathway. Rendering the LI motif nonfunctional inhibited gap junction formation by augmenting the endocytosis of Cx32 via the LL and LR motifs. Our studies have defined distinct roles of these motifs in regulating the endocytosis of Cx32 and its gap junction-forming ability.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Anuttoma Ray
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Parul Katoch
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Nimansha Jain
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Parmender P Mehta
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
59
|
Protein Kinase C Enhances Electrical Synaptic Transmission by Acting on Junctional and Postsynaptic Ca 2+ Currents. J Neurosci 2018; 38:2796-2808. [PMID: 29440551 DOI: 10.1523/jneurosci.2619-17.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 01/15/2018] [Accepted: 02/02/2018] [Indexed: 11/21/2022] Open
Abstract
By synchronizing neuronal activity, electrical transmission influences the coordination, pattern, and/or frequency of firing. In the hemaphroditic marine-snail, Aplysia calfornica, the neuroendocrine bag cell neurons use electrical synapses to synchronize a 30 min afterdischarge of action potentials for the release of reproductive hormone. During the afterdischarge, protein kinase C (PKC) is activated, although its impact on bag cell neuron electrical transmission is unknown. This was investigated here by monitoring electrical synapses between paired cultured bag cell neurons using dual whole-cell recording. Voltage clamp revealed a largely voltage-independent junctional current, which was enhanced by treating with a PKC activator, PMA, before recording. We also examined the transfer of presynaptic action potential-like waveforms (generated in voltage clamp) to the postsynaptic cell (measured in current clamp). For control pairs, the presynaptic spike-like waveforms mainly evoked electrotonic potentials; however, when PKC was triggered, these stimuli consistently produced postsynaptic action potentials. To assess whether this involved changes to postsynaptic responsiveness, single bag cell neurons were injected with junctional-like current mimicking that evoked by a presynaptic action potential. Unlike control neurons, which were less likely to spike, cells in PMA always fired action potentials to the junctional-like current. Furthermore, PKC activation increased a postsynaptic voltage-gated Ca2+ current, which was recruited even by modest depolarization associated with an electrotonic potential. Whereas PKC inhibits gap junctions in most systems, bag cell neurons are rather unique, as the kinase potentiates the electrical synapse; in turn, this synergizes with augmented postsynaptic Ca2+ current to promote synchronous firing.SIGNIFICANCE STATEMENT Electrical coupling is a fundamental form of communication. For the bag cell neurons of Aplysia, electrical synapses coordinate a prolonged burst of action potentials known as the afterdischarge. We looked at how protein kinase C, which is upregulated with the afterdischarge, influences information transfer across the synapse. The kinase activation increased junctional current, a remarkable finding given that this enzyme is largely considered inhibitory for gap junctions. There was also an augmentation in the ability of a presynaptic neuron to provoke postsynaptic action potentials. This increased excitability was, in part, due to enhanced postsynaptic voltage-dependent Ca2+ current. Thus, protein kinase C improves the fidelity of electrotonic transmission and promotes synchronous firing by modulating both junctional and membrane conductances.
Collapse
|
60
|
Abstract
Astrocytes are neural cells of ectodermal, neuroepithelial origin that provide for homeostasis and defense of the central nervous system (CNS). Astrocytes are highly heterogeneous in morphological appearance; they express a multitude of receptors, channels, and membrane transporters. This complement underlies their remarkable adaptive plasticity that defines the functional maintenance of the CNS in development and aging. Astrocytes are tightly integrated into neural networks and act within the context of neural tissue; astrocytes control homeostasis of the CNS at all levels of organization from molecular to the whole organ.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| | - Maiken Nedergaard
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| |
Collapse
|
61
|
Verkhratsky A, Nedergaard M. Physiology of Astroglia. Physiol Rev 2018; 98:239-389. [PMID: 29351512 PMCID: PMC6050349 DOI: 10.1152/physrev.00042.2016] [Citation(s) in RCA: 964] [Impact Index Per Article: 160.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/22/2017] [Accepted: 04/27/2017] [Indexed: 02/07/2023] Open
Abstract
Astrocytes are neural cells of ectodermal, neuroepithelial origin that provide for homeostasis and defense of the central nervous system (CNS). Astrocytes are highly heterogeneous in morphological appearance; they express a multitude of receptors, channels, and membrane transporters. This complement underlies their remarkable adaptive plasticity that defines the functional maintenance of the CNS in development and aging. Astrocytes are tightly integrated into neural networks and act within the context of neural tissue; astrocytes control homeostasis of the CNS at all levels of organization from molecular to the whole organ.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| | - Maiken Nedergaard
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| |
Collapse
|
62
|
Calkins TL, Piermarini PM. A Blood Meal Enhances Innexin mRNA Expression in the Midgut, Malpighian Tubules, and Ovaries of the Yellow Fever Mosquito Aedes aegypti. INSECTS 2017; 8:insects8040122. [PMID: 29113099 PMCID: PMC5746805 DOI: 10.3390/insects8040122] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 10/30/2017] [Accepted: 10/31/2017] [Indexed: 11/16/2022]
Abstract
Mosquitoes are vectors of pathogens that cause diseases of medical and veterinary importance. Female mosquitoes transmit these pathogens while taking a blood meal, which most species require to produce eggs. The period after a blood meal is a time of extreme physiological change that requires rapid coordination of specific tissues. Gap junctions (GJ) are intercellular channels that aid in the coordination of cells within tissues via the direct transfer of certain small molecules and ions between cells. Evolutionarily distinct groups of proteins form the gap junctions of vertebrate and invertebrate animals (connexins and innexins, respectively). Aedes aegypti mosquitoes possess six genes encoding innexins: inx1, inx2, inx3, inx4, inx7, and inx8. The goal of this study was to identify potential roles of innexins in the physiology of mosquitoes after a blood meal by using qPCR to quantify their mRNA expression in adult females at 3 h and 24 h post-blood meal (PBM) relative to non-blood-fed controls. We found that at 24 h PBM, expression levels of inx2, inx3, and inx4 mRNAs increased; inx2 was the most highly upregulated innexin in key tissues associated with blood-meal digestion and egg production (i.e., the midgut and ovaries, respectively). However, knocking down inx2 mRNA levels by over 75% via RNA interference had no significant effect on fecundity. Altogether, our results suggest that a blood meal influences the molecular expression of innexins in mosquitoes, but their specific physiological roles remain to be elucidated.
Collapse
Affiliation(s)
- Travis L Calkins
- Department of Entomology, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA.
| | - Peter M Piermarini
- Department of Entomology, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA.
| |
Collapse
|
63
|
Haefliger JA, Allagnat F, Hamard L, Le Gal L, Meda P, Nardelli-Haefliger D, Génot E, Alonso F. Targeting Cx40 (Connexin40) Expression or Function Reduces Angiogenesis in the Developing Mouse Retina. Arterioscler Thromb Vasc Biol 2017; 37:2136-2146. [PMID: 28982669 DOI: 10.1161/atvbaha.117.310072] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 09/20/2017] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Cx40 (Connexin40) forms intercellular channels that coordinate the electric conduction in the heart and the vasomotor tone in large vessels. The protein was shown to regulate tumoral angiogenesis; however, whether Cx40 also contributes to physiological angiogenesis is still unknown. APPROACH AND RESULTS Here, we show that Cx40 contributes to physiological angiogenesis. Genetic deletion of Cx40 leads to a reduction in vascular growth and capillary density in the neovascularization model of the mouse neonatal retina. At the angiogenic front, vessel sprouting is reduced, and the mural cells recruited along the sprouts display an altered phenotype. These alterations can be attributed to disturbed endothelial cell functions as selective reexpression of Cx40 in these cells restores normal angiogenesis. In vitro, targeting Cx40 in microvascular endothelial cells, by silencing its expression or by blocking gap junction channels, decreases their proliferation. Moreover, loss of Cx40 in these cells also increases their release of PDGF (platelet-derived growth factor) and promotes the chemoattraction of mural cells. In vivo, an intravitreal injection of a Cx40 inhibitory peptide, phenocopies the loss of Cx40 in the retinal vasculature of wild-type mice. CONCLUSIONS Collectively, our data show that endothelial Cx40 contributes to the early stages of physiological angiogenesis in the developing retina, by regulating vessel growth and maturation. Cx40 thus represents a novel therapeutic target for treating pathological ocular angiogenesis.
Collapse
Affiliation(s)
- Jacques-Antoine Haefliger
- From the Department of Medicine (J.-A.H., F.A., L.H., L.L.G., F.A.) and Department of Urology (D.N.H.), Lausanne University Hospital, Switzerland; Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Switzerland (P.M.); and Centre de Recherche Cardio-Thoracique de Bordeaux (INSERM U1045), Université de Bordeaux, France (E.G., F.A.).
| | - Florent Allagnat
- From the Department of Medicine (J.-A.H., F.A., L.H., L.L.G., F.A.) and Department of Urology (D.N.H.), Lausanne University Hospital, Switzerland; Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Switzerland (P.M.); and Centre de Recherche Cardio-Thoracique de Bordeaux (INSERM U1045), Université de Bordeaux, France (E.G., F.A.)
| | - Lauriane Hamard
- From the Department of Medicine (J.-A.H., F.A., L.H., L.L.G., F.A.) and Department of Urology (D.N.H.), Lausanne University Hospital, Switzerland; Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Switzerland (P.M.); and Centre de Recherche Cardio-Thoracique de Bordeaux (INSERM U1045), Université de Bordeaux, France (E.G., F.A.)
| | - Loïc Le Gal
- From the Department of Medicine (J.-A.H., F.A., L.H., L.L.G., F.A.) and Department of Urology (D.N.H.), Lausanne University Hospital, Switzerland; Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Switzerland (P.M.); and Centre de Recherche Cardio-Thoracique de Bordeaux (INSERM U1045), Université de Bordeaux, France (E.G., F.A.)
| | - Paolo Meda
- From the Department of Medicine (J.-A.H., F.A., L.H., L.L.G., F.A.) and Department of Urology (D.N.H.), Lausanne University Hospital, Switzerland; Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Switzerland (P.M.); and Centre de Recherche Cardio-Thoracique de Bordeaux (INSERM U1045), Université de Bordeaux, France (E.G., F.A.)
| | - Denise Nardelli-Haefliger
- From the Department of Medicine (J.-A.H., F.A., L.H., L.L.G., F.A.) and Department of Urology (D.N.H.), Lausanne University Hospital, Switzerland; Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Switzerland (P.M.); and Centre de Recherche Cardio-Thoracique de Bordeaux (INSERM U1045), Université de Bordeaux, France (E.G., F.A.)
| | - Elisabeth Génot
- From the Department of Medicine (J.-A.H., F.A., L.H., L.L.G., F.A.) and Department of Urology (D.N.H.), Lausanne University Hospital, Switzerland; Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Switzerland (P.M.); and Centre de Recherche Cardio-Thoracique de Bordeaux (INSERM U1045), Université de Bordeaux, France (E.G., F.A.)
| | - Florian Alonso
- From the Department of Medicine (J.-A.H., F.A., L.H., L.L.G., F.A.) and Department of Urology (D.N.H.), Lausanne University Hospital, Switzerland; Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Switzerland (P.M.); and Centre de Recherche Cardio-Thoracique de Bordeaux (INSERM U1045), Université de Bordeaux, France (E.G., F.A.).
| |
Collapse
|
64
|
Umrani MR, Joglekar MV, Somerville Glover E, Wong W, Hardikar AA. Connexins and microRNAs: Interlinked players in regulating islet function? Islets 2017; 9:99-108. [PMID: 28686518 PMCID: PMC5624287 DOI: 10.1080/19382014.2017.1331192] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 05/10/2017] [Accepted: 05/11/2017] [Indexed: 02/09/2023] Open
Abstract
Pancreatic β-cells are connected to neighboring endocrine cells through the adherin proteins and gap junctions. Connexin 36 (Cx36) is one of the most well-studied and abundantly expressed gap-junction proteins within rodent islets, which is important in coordinated insulin secretion. The expression of connexins is regulated at various levels and by several mechanisms; one of which is via microRNAs. In past 2 decades, microRNAs (miRNAs) have emerged as key molecules in developmental, physiologic and pathological processes. However, very few studies have demonstrated miRNA-mediated regulation of connexins. Even though there are no reports yet on miRNAs and Cx36; we envisage that considering the important role of connexins and microRNAs in insulin secretion, there would be common pathways interlinking these biomolecules. Here, we discuss the current literature on connexins and miRNAs specifically with reference to islet function.
Collapse
Affiliation(s)
- Malati R. Umrani
- National centre for cell science, Ganeshkhind, Pune University Campus, Pune, India
- Diabetes and Islet Biology Group, NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - Mugdha V. Joglekar
- Diabetes and Islet Biology Group, NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - Ella Somerville Glover
- Diabetes and Islet Biology Group, NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - Wilson Wong
- Diabetes and Islet Biology Group, NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - Anandwardhan A. Hardikar
- Diabetes and Islet Biology Group, NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia
| |
Collapse
|
65
|
Boucher J, Monvoisin A, Vix J, Mesnil M, Thuringer D, Debiais F, Cronier L. Connexins, important players in the dissemination of prostate cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:202-215. [PMID: 28693897 DOI: 10.1016/j.bbamem.2017.06.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 06/22/2017] [Accepted: 06/29/2017] [Indexed: 12/25/2022]
Abstract
Over the past 50years, increasing experimental evidences have established that connexins (Cxs) and gap junctional intercellular communication (GJIC) ensure an important role in both the onset and development of cancerous processes. In the present review, we focus on the impact of Cxs and GJIC during the development of prostate cancer (PCa), from the primary growth mainly localized in acinar glands and ducts to the distant metastasis mainly concentrated in bone. As observed in several other types of solid tumours, Cxs and especially Cx43 exhibit an ambivalent role with a tumour suppressor effect in the early stages and, conversely, a rather pro-tumoural profile for most of invasion and dissemination steps to secondary sites. We report here the current knowledge on the function of Cxs during PCa cells migration, cytoskeletal dynamics, proteinases activities and the cross talk with the surrounding stromal cells in the microenvironment of the tumour and the bones. In addition, we discuss the role of Cxs in the bone tropism even if the prostate model is rarely used to study the complete sequence of cancer dissemination compared to breast cancer or melanoma. Even if not yet fully understood, these recent findings on Cxs provide new insights into their molecular mechanisms associated with progression and bone targeted behaviour of PCa. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Jonathan Boucher
- Laboratory Signalisation et Transports Ioniques Membranaires (STIM), ERL7368 - CNRS, University of Poitiers, Poitiers, France
| | - Arnaud Monvoisin
- Laboratory Signalisation et Transports Ioniques Membranaires (STIM), ERL7368 - CNRS, University of Poitiers, Poitiers, France
| | - Justine Vix
- Laboratory Signalisation et Transports Ioniques Membranaires (STIM), ERL7368 - CNRS, University of Poitiers, Poitiers, France; Department of Rheumatology, C.H.U. la Milétrie, Poitiers, France
| | - Marc Mesnil
- Laboratory Signalisation et Transports Ioniques Membranaires (STIM), ERL7368 - CNRS, University of Poitiers, Poitiers, France
| | | | - Françoise Debiais
- Laboratory Signalisation et Transports Ioniques Membranaires (STIM), ERL7368 - CNRS, University of Poitiers, Poitiers, France; Department of Rheumatology, C.H.U. la Milétrie, Poitiers, France
| | - Laurent Cronier
- Laboratory Signalisation et Transports Ioniques Membranaires (STIM), ERL7368 - CNRS, University of Poitiers, Poitiers, France.
| |
Collapse
|
66
|
Wang SG, Tsao DD, Vanderpool KG, Yasumura T, Rash JE, Nagy JI. Connexin36 localization to pinealocytes in the pineal gland of mouse and rat. Eur J Neurosci 2017; 45:1594-1605. [PMID: 28474748 PMCID: PMC5507615 DOI: 10.1111/ejn.13602] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 04/27/2017] [Accepted: 04/28/2017] [Indexed: 01/07/2023]
Abstract
Several cell types in the pineal gland are known to establish intercellular gap junctions, but the connexin constituents of those junctions have not been fully characterized. Specifically, the expression of connexin36 (Cx36) protein and mRNA has been examined in the pineal, but the identity of cells that produce Cx36 and that form Cx36-containing gap junctions has not been determined. We used immunofluorescence and freeze fracture replica immunogold labelling (FRIL) of Cx36 to investigate the cellular and subcellular localization of Cx36 in the pineal gland of adult mouse and rat. Immunofluorescence labelling of Cx36 was visualized exclusively as puncta or short immunopositive strands that were distributed throughout the pineal, and which were absent in pineal sections from Cx36 null mice. By double immunofluorescence labelling, Cx36 was localized to tryptophan hydroxylase-positive and 5-hydroxytryptamine-positive pinealocyte cell bodies and their large initial processes, including at intersections of those processes and at sites displaying a confluence of processes. Labelling for the cell junction marker zonula occludens-1 (ZO-1) either overlapped or was closely associated with labelling for Cx36. Pinealocytes thus form Cx36-containing gap junctions that also incorporate the scaffolding protein ZO-1. FRIL revealed labelling of Cx36 at ultrastructurally defined gap junctions between pinealocytes, most of which was at gap junctions having reticular, ribbon or string configurations. The results suggest that the endocrine functions of pinealocytes and their secretion of melatonin is supported by their intercellular communication via Cx36-containing gap junctions, which may now be tested by the use of Cx36 null mice.
Collapse
Affiliation(s)
- S G Wang
- Department of Physiology, Faculty of Medicine, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB, R3E 0J9, Canada
| | - D D Tsao
- Department of Physiology, Faculty of Medicine, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB, R3E 0J9, Canada
| | - K G Vanderpool
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - T Yasumura
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - J E Rash
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - J I Nagy
- Department of Physiology, Faculty of Medicine, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB, R3E 0J9, Canada
| |
Collapse
|
67
|
Meda P. Gap junction proteins are key drivers of endocrine function. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:124-140. [PMID: 28284720 DOI: 10.1016/j.bbamem.2017.03.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/03/2017] [Accepted: 03/06/2017] [Indexed: 01/07/2023]
Abstract
It has long been known that the main secretory cells of exocrine and endocrine glands are connected by gap junctions, made by a variety of connexin species that ensure their electrical and metabolic coupling. Experiments in culture systems and animal models have since provided increasing evidence that connexin signaling contributes to control the biosynthesis and release of secretory products, as well as to the life and death of secretory cells. More recently, genetic studies have further provided the first lines of evidence that connexins also control the function of human glands, which are central to the pathogenesis of major endocrine diseases. Here, we summarize the recent information gathered on connexin signaling in these systems, since the last reviews on the topic, with particular regard to the pancreatic beta cells which produce insulin, and the renal cells which produce renin. These cells are keys to the development of various forms of diabetes and hypertension, respectively, and combine to account for the exploding, worldwide prevalence of the metabolic syndrome. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva Medical School, Switzerland.
| |
Collapse
|
68
|
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
|
69
|
Striegel DA, Hara M, Periwal V. Adaptation of pancreatic islet cyto-architecture during development. Phys Biol 2016; 13:025004. [PMID: 27063927 DOI: 10.1088/1478-3975/13/2/025004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Plasma glucose in mammals is regulated by hormones secreted by the islets of Langerhans embedded in the exocrine pancreas. Islets consist of endocrine cells, primarily α, β, and δ cells, which secrete glucagon, insulin, and somatostatin, respectively. β cells form irregular locally connected clusters within islets that act in concert to secrete insulin upon glucose stimulation. Varying demands and available nutrients during development produce changes in the local connectivity of β cells in an islet. We showed in earlier work that graph theory provides a framework for the quantification of the seemingly stochastic cyto-architecture of β cells in an islet. To quantify the dynamics of endocrine connectivity during development requires a framework for characterizing changes in the probability distribution on the space of possible graphs, essentially a Fokker-Planck formalism on graphs. With large-scale imaging data for hundreds of thousands of islets containing millions of cells from human specimens, we show that this dynamics can be determined quantitatively. Requiring that rearrangement and cell addition processes match the observed dynamic developmental changes in quantitative topological graph characteristics strongly constrained possible processes. Our results suggest that there is a transient shift in preferred connectivity for β cells between 1-35 weeks and 12-24 months.
Collapse
|
70
|
Alonso F, Domingos-Pereira S, Le Gal L, Derré L, Meda P, Jichlinski P, Nardelli-Haefliger D, Haefliger JA. Targeting endothelial connexin40 inhibits tumor growth by reducing angiogenesis and improving vessel perfusion. Oncotarget 2016; 7:14015-28. [PMID: 26883111 PMCID: PMC4924695 DOI: 10.18632/oncotarget.7370] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 01/29/2016] [Indexed: 01/09/2023] Open
Abstract
Endothelial connexin40 (Cx40) contributes to regulate the structure and function of vessels. We have examined whether the protein also modulates the altered growth of vessels in tumor models established in control mice (WT), mice lacking Cx40 (Cx40-/-), and mice expressing the protein solely in endothelial cells (Tie2-Cx40). Tumoral angiogenesis and growth were reduced, whereas vessel perfusion, smooth muscle cell (SMC) coverage and animal survival were increased in Cx40-/- but not Tie2-Cx40 mice, revealing a critical involvement of endothelial Cx40 in transformed tissues independently of the hypertensive status of Cx40-/- mice. As a result, Cx40-/- mice bearing tumors survived significantly longer than corresponding controls, including after a cytotoxic administration. Comparable observations were made in WT mice injected with a peptide targeting Cx40, supporting the Cx40 involvement. This involvement was further confirmed in the absence of Cx40 or by peptide-inhibition of this connexin in aorta-sprouting, matrigel plug and SMC migration assays, and associated with a decreased expression of the phosphorylated form of endothelial nitric oxide synthase. The data identify Cx40 as a potential novel target in cancer treatment.
Collapse
MESH Headings
- Animals
- Aorta/pathology
- Apoptosis
- Biomarkers, Tumor/metabolism
- Blood Vessels/physiology
- Cell Proliferation
- Connexins/antagonists & inhibitors
- Connexins/metabolism
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Female
- Humans
- Lung Neoplasms/blood supply
- Lung Neoplasms/pathology
- Lung Neoplasms/prevention & control
- Melanoma, Experimental/blood supply
- Melanoma, Experimental/pathology
- Melanoma, Experimental/prevention & control
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasm Invasiveness
- Neovascularization, Pathologic/prevention & control
- Perfusion
- Tumor Cells, Cultured
- Urinary Bladder Neoplasms/blood supply
- Urinary Bladder Neoplasms/pathology
- Urinary Bladder Neoplasms/prevention & control
- Gap Junction alpha-5 Protein
Collapse
Affiliation(s)
- Florian Alonso
- Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | | | - Loïc Le Gal
- Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Laurent Derré
- Department of Urology, Lausanne University Hospital, Lausanne, Switzerland
| | - Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva, Medical Center, Geneva, Switzerland
| | - Patrice Jichlinski
- Department of Urology, Lausanne University Hospital, Lausanne, Switzerland
| | | | | |
Collapse
|
71
|
Cigliola V, Populaire C, Pierri CL, Deutsch S, Haefliger JA, Fadista J, Lyssenko V, Groop L, Rueedi R, Thorel F, Herrera PL, Meda P. A Variant of GJD2, Encoding for Connexin 36, Alters the Function of Insulin Producing β-Cells. PLoS One 2016; 11:e0150880. [PMID: 26959991 PMCID: PMC4784816 DOI: 10.1371/journal.pone.0150880] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/20/2016] [Indexed: 01/16/2023] Open
Abstract
Signalling through gap junctions contributes to control insulin secretion and, thus, blood glucose levels. Gap junctions of the insulin-producing β-cells are made of connexin 36 (Cx36), which is encoded by the GJD2 gene. Cx36-null mice feature alterations mimicking those observed in type 2 diabetes (T2D). GJD2 is also expressed in neurons, which share a number of common features with pancreatic β-cells. Given that a synonymous exonic single nucleotide polymorphism of human Cx36 (SNP rs3743123) associates with altered function of central neurons in a subset of epileptic patients, we investigated whether this SNP also caused alterations of β-cell function. Transfection of rs3743123 cDNA in connexin-lacking HeLa cells resulted in altered formation of gap junction plaques and cell coupling, as compared to those induced by wild type (WT) GJD2 cDNA. Transgenic mice expressing the very same cDNAs under an insulin promoter revealed that SNP rs3743123 expression consistently lead to a post-natal reduction of islet Cx36 levels and β-cell survival, resulting in hyperglycemia in selected lines. These changes were not observed in sex- and age-matched controls expressing WT hCx36. The variant GJD2 only marginally associated to heterogeneous populations of diabetic patients. The data document that a silent polymorphism of GJD2 is associated with altered β-cell function, presumably contributing to T2D pathogenesis.
Collapse
Affiliation(s)
- Valentina Cigliola
- Department of Genetic Medicine and Development, University of Geneva Faculty of Medicine, Geneva, Switzerland
| | - Celine Populaire
- Centre Hospitalier Régional Universitaire Besançon, Besançon, France
| | - Ciro L. Pierri
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Samuel Deutsch
- Joint Genome Institute, Walnut Creek, California, United States of America
| | | | - João Fadista
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University, Malmö, Sweden
| | - Valeriya Lyssenko
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University, Malmö, Sweden
- Steno Diabetes Center A/S, Gentofte, Denmark
| | - Leif Groop
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University, Malmö, Sweden
| | - Rico Rueedi
- Department of Computational Biology, University of Lausanne, Rue du Bugnon 27, 1011, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, 1015, Lausanne, Switzerland
| | - Fabrizio Thorel
- Department of Genetic Medicine and Development, University of Geneva Faculty of Medicine, Geneva, Switzerland
| | - Pedro Luis Herrera
- Department of Genetic Medicine and Development, University of Geneva Faculty of Medicine, Geneva, Switzerland
| | - Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva Faculty of Medicine, Geneva, Switzerland
| |
Collapse
|
72
|
Marsh A, Casey-Green K, Probert F, Withall D, Mitchell DA, Dilly SJ, James S, Dimitri W, Ladwa SR, Taylor PC, Singer DRJ. Simvastatin Sodium Salt and Fluvastatin Interact with Human Gap Junction Gamma-3 Protein. PLoS One 2016; 11:e0148266. [PMID: 26863535 PMCID: PMC4749215 DOI: 10.1371/journal.pone.0148266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 01/15/2016] [Indexed: 11/26/2022] Open
Abstract
Finding pleiomorphic targets for drugs allows new indications or warnings for treatment to be identified. As test of concept, we applied a new chemical genomics approach to uncover additional targets for the widely prescribed lipid-lowering pro-drug simvastatin. We used mRNA extracted from internal mammary artery from patients undergoing coronary artery surgery to prepare a viral cardiovascular protein library, using T7 bacteriophage. We then studied interactions of clones of the bacteriophage, each expressing a different cardiovascular polypeptide, with surface-bound simvastatin in 96-well plates. To maximise likelihood of identifying meaningful interactions between simvastatin and vascular peptides, we used a validated photo-immobilisation method to apply a series of different chemical linkers to bind simvastatin so as to present multiple orientations of its constituent components to potential targets. Three rounds of biopanning identified consistent interaction with the clone expressing part of the gene GJC3, which maps to Homo sapiens chromosome 7, and codes for gap junction gamma-3 protein, also known as connexin 30.2/31.3 (mouse connexin Cx29). Further analysis indicated the binding site to be for the N-terminal domain putatively 'regulating' connexin hemichannel and gap junction pores. Using immunohistochemistry we found connexin 30.2/31.3 to be present in samples of artery similar to those used to prepare the bacteriophage library. Surface plasmon resonance revealed that a 25 amino acid synthetic peptide representing the discovered N-terminus did not interact with simvastatin lactone, but did bind to the hydrolysed HMG CoA inhibitor, simvastatin acid. This interaction was also seen for fluvastatin. The gap junction blockers carbenoxolone and flufenamic acid also interacted with the same peptide providing insight into potential site of binding. These findings raise key questions about the functional significance of GJC3 transcripts in the vasculature and other tissues, and this connexin's role in therapeutic and adverse effects of statins in a range of disease states.
Collapse
Affiliation(s)
- Andrew Marsh
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | | | - Fay Probert
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - David Withall
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - Daniel A. Mitchell
- Division of Metabolic and Vascular Health, Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, Coventry, CV2 2DX, United Kingdom
| | - Suzanne J. Dilly
- Tangent Reprofiling Ltd, c/o SEEK, Central Point, 45 Beech Street, London, EC2Y 8AD, United Kingdom
| | - Sean James
- University Hospital Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, United Kingdom
| | - Wade Dimitri
- University Hospital Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, United Kingdom
| | - Sweta R. Ladwa
- Tangent Reprofiling Ltd, c/o SEEK, Central Point, 45 Beech Street, London, EC2Y 8AD, United Kingdom
| | - Paul C. Taylor
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - Donald R. J. Singer
- University Hospital Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, United Kingdom
- Fellowship of Postgraduate Medicine, 11 Chandos St, London W1G 9EB, United Kingdom
- Division of Metabolic and Vascular Health, Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, Coventry, CV2 2DX, United Kingdom
| |
Collapse
|
73
|
Sala G, Badalamenti S, Ponticelli C. The Renal Connexome and Possible Roles of Connexins in Kidney Diseases. Am J Kidney Dis 2015; 67:677-87. [PMID: 26613807 DOI: 10.1053/j.ajkd.2015.09.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 09/30/2015] [Indexed: 12/21/2022]
Abstract
Connexins are membrane-spanning proteins that allow for the formation of cell-to-cell channels and cell-to-extracellular space hemichannels. Many connexin subtypes are expressed in kidney cells. Some mutations in connexin genes have been linked to various human pathologies, including cardiovascular, neurodegenerative, lung, and skin diseases, but the exact role of connexins in kidney disease remains unclear. Some hypotheses about a connection between genetic mutations, endoplasmic reticulum (ER) stress, and the unfolded protein response (UPR) in kidney pathology have been explored. The potential relationship of kidney disease to abnormal production of connexin proteins, mutations in their genes together with ER stress, or the UPR is still a matter of debate. In this scenario, it is tantalizing to speculate about a possible role of connexins in the setting of kidney pathologies that are thought to be caused by a deregulated podocyte protein expression, the so-called podocytopathies. In this article, we give examples of the roles of connexins in kidney (patho)physiology and propose avenues for further research concerning connexins, ER stress, and UPR in podocytopathies that may ultimately help refine drug treatment.
Collapse
Affiliation(s)
- Gabriele Sala
- Nephrology and Dialysis Unit, Humanitas Clinical Research Center, Rozzano (Milano), Italy.
| | - Salvatore Badalamenti
- Nephrology and Dialysis Unit, Humanitas Clinical Research Center, Rozzano (Milano), Italy
| | - Claudio Ponticelli
- Nephrology and Dialysis Unit, Humanitas Clinical Research Center, Rozzano (Milano), Italy
| |
Collapse
|
74
|
Abstract
The pancreas produces enzymes with a digestive function and hormones with a metabolic function, which are produced by distinct cell types of acini and islets, respectively. Within these units, secretory cells coordinate their functioning by exchanging information via signals that flow in the intercellular spaces and are generated either at distance (several neural and hormonal inputs) or nearby the pancreatic cells themselves (inputs mediated by membrane ionic-specific channels and by ionic- and metabolite-permeant pannexin channels and connexin "hemichannels"). Pancreatic secretory cells further interact via the extracellular matrix of the pancreas (inputs mediated by integrins) and directly with neighboring cells, by mechanisms that do not require extracellular mediators (inputs mediated by gap and tight junction channels). Here, we review the expression and function of the connexins and pannexins that are expressed by the main secretory cells of the exocrine and endocrine pancreatic cells. Available data show that the patterns of expression of these proteins differ in acini and islets, supporting distinct functions in the physiological secretion of pancreatic enzymes and hormones. Circumstantial evidence further suggests that alterations in the signaling provided by these proteins are involved in pancreatic diseases.
Collapse
|
75
|
Dolenšek J, Špelič D, Skelin Klemen M, Žalik B, Gosak M, Slak Rupnik M, Stožer A. Membrane Potential and Calcium Dynamics in Beta Cells from Mouse Pancreas Tissue Slices: Theory, Experimentation, and Analysis. SENSORS 2015; 15:27393-419. [PMID: 26516866 PMCID: PMC4701238 DOI: 10.3390/s151127393] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 10/11/2015] [Accepted: 10/14/2015] [Indexed: 12/17/2022]
Abstract
Beta cells in the pancreatic islets of Langerhans are precise biological sensors for glucose and play a central role in balancing the organism between catabolic and anabolic needs. A hallmark of the beta cell response to glucose are oscillatory changes of membrane potential that are tightly coupled with oscillatory changes in intracellular calcium concentration which, in turn, elicit oscillations of insulin secretion. Both membrane potential and calcium changes spread from one beta cell to the other in a wave-like manner. In order to assess the properties of the abovementioned responses to physiological and pathological stimuli, the main challenge remains how to effectively measure membrane potential and calcium changes at the same time with high spatial and temporal resolution, and also in as many cells as possible. To date, the most wide-spread approach has employed the electrophysiological patch-clamp method to monitor membrane potential changes. Inherently, this technique has many advantages, such as a direct contact with the cell and a high temporal resolution. However, it allows one to assess information from a single cell only. In some instances, this technique has been used in conjunction with CCD camera-based imaging, offering the opportunity to simultaneously monitor membrane potential and calcium changes, but not in the same cells and not with a reliable cellular or subcellular spatial resolution. Recently, a novel family of highly-sensitive membrane potential reporter dyes in combination with high temporal and spatial confocal calcium imaging allows for simultaneously detecting membrane potential and calcium changes in many cells at a time. Since the signals yielded from both types of reporter dyes are inherently noisy, we have developed complex methods of data denoising that permit for visualization and pixel-wise analysis of signals. Combining the experimental approach of high-resolution imaging with the advanced analysis of noisy data enables novel physiological insights and reassessment of current concepts in unprecedented detail.
Collapse
Affiliation(s)
- Jurij Dolenšek
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; E-Mails: (J.D.); (M.S.K.); (M.G.); (M.S.R.)
| | - Denis Špelič
- Faculty of Electrical Engineering and Computer Science, University of Maribor, SI-2000 Maribor, Slovenia; E-Mails: (D.Š.); (B.Ž.)
| | - Maša Skelin Klemen
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; E-Mails: (J.D.); (M.S.K.); (M.G.); (M.S.R.)
| | - Borut Žalik
- Faculty of Electrical Engineering and Computer Science, University of Maribor, SI-2000 Maribor, Slovenia; E-Mails: (D.Š.); (B.Ž.)
- Center for Open Innovation and Research, Core@UM, University of Maribor, SI-2000 Maribor, Slovenia
| | - Marko Gosak
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; E-Mails: (J.D.); (M.S.K.); (M.G.); (M.S.R.)
- Center for Open Innovation and Research, Core@UM, University of Maribor, SI-2000 Maribor, Slovenia
- Department of Physics, Faculty of Natural Sciences and Mathematics, University of Maribor, SI-2000 Maribor, Slovenia
| | - Marjan Slak Rupnik
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; E-Mails: (J.D.); (M.S.K.); (M.G.); (M.S.R.)
- Center for Open Innovation and Research, Core@UM, University of Maribor, SI-2000 Maribor, Slovenia
- Center for Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Andraž Stožer
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; E-Mails: (J.D.); (M.S.K.); (M.G.); (M.S.R.)
- Center for Open Innovation and Research, Core@UM, University of Maribor, SI-2000 Maribor, Slovenia
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +386-2-2345843
| |
Collapse
|
76
|
Loppini A, Braun M, Filippi S, Pedersen MG. Mathematical modeling of gap junction coupling and electrical activity in human β-cells. Phys Biol 2015; 12:066002. [PMID: 26403477 DOI: 10.1088/1478-3975/12/6/066002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Coordinated insulin secretion is controlled by electrical coupling of pancreatic β-cells due to connexin-36 gap junctions. Gap junction coupling not only synchronizes the heterogeneous β-cell population, but can also modify the electrical behavior of the cells. These phenomena have been widely studied with mathematical models based on data from mouse β-cells. However, it is now known that human β-cell electrophysiology shows important differences to its rodent counterpart, and although human pancreatic islets express connexin-36 and show evidence of β-cell coupling, these aspects have been little investigated in human β-cells. Here we investigate theoretically, the gap junction coupling strength required for synchronizing electrical activity in a small cluster of cells simulated with a recent mathematical model of human β-cell electrophysiology. We find a lower limit for the coupling strength of approximately 20 pS (i.e., normalized to cell size, ∼2 pS pF(-1)) below which spiking electrical activity is asynchronous. To confront this theoretical lower bound with data, we use our model to estimate from an experimental patch clamp recording that the coupling strength is approximately 100-200 pS (10-20 pS pF(-1)), similar to previous estimates in mouse β-cells. We then investigate the role of gap junction coupling in synchronizing and modifying other forms of electrical activity in human β-cell clusters. We find that electrical coupling can prolong the period of rapid bursting electrical activity, and synchronize metabolically driven slow bursting, in particular when the metabolic oscillators are in phase. Our results show that realistic coupling conductances are sufficient to promote synchrony in small clusters of human β-cells as observed experimentally, and provide motivation for further detailed studies of electrical coupling in human pancreatic islets.
Collapse
Affiliation(s)
- Alessandro Loppini
- Nonlinear Physics and Mathematical Modeling Laboratory, University Campus Bio-Medico, I-00128, Rome, Italy
| | | | | | | |
Collapse
|
77
|
Striegel DA, Hara M, Periwal V. The Beta Cell in Its Cluster: Stochastic Graphs of Beta Cell Connectivity in the Islets of Langerhans. PLoS Comput Biol 2015; 11:e1004423. [PMID: 26266953 PMCID: PMC4534467 DOI: 10.1371/journal.pcbi.1004423] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 07/02/2015] [Indexed: 12/25/2022] Open
Abstract
Pancreatic islets of Langerhans consist of endocrine cells, primarily α, β and δ cells, which secrete glucagon, insulin, and somatostatin, respectively, to regulate plasma glucose. β cells form irregular locally connected clusters within islets that act in concert to secrete insulin upon glucose stimulation. Due to the central functional significance of this local connectivity in the placement of β cells in an islet, it is important to characterize it quantitatively. However, quantification of the seemingly stochastic cytoarchitecture of β cells in an islet requires mathematical methods that can capture topological connectivity in the entire β-cell population in an islet. Graph theory provides such a framework. Using large-scale imaging data for thousands of islets containing hundreds of thousands of cells in human organ donor pancreata, we show that quantitative graph characteristics differ between control and type 2 diabetic islets. Further insight into the processes that shape and maintain this architecture is obtained by formulating a stochastic theory of β-cell rearrangement in whole islets, just as the normal equilibrium distribution of the Ornstein-Uhlenbeck process can be viewed as the result of the interplay between a random walk and a linear restoring force. Requiring that rearrangements maintain the observed quantitative topological graph characteristics strongly constrained possible processes. Our results suggest that β-cell rearrangement is dependent on its connectivity in order to maintain an optimal cluster size in both normal and T2D islets. High or low blood glucose levels are detrimental to human health. The hormone-secreting cells primarily responsible for maintaining glucose at physiologically appropriate levels are embedded in small clusters within the pancreas, the so-called islets of Langerhans. These islets have an irregular arrangement of cells, β cells that secrete insulin, α cells that secrete glucagon, and other cells with less well-understood functions. While the arrangement of β cells is irregular, these cells need to be touching for the islet to respond to glucose with insulin secretion. We first use a mathematical formalism called graph theory to show that cell arrangements in islets from diabetic and control donors are significantly different. The question we then address is: Is there some set of moves of islet cells that will preserve the observed arrangement? The aim is to gain insight into the biological processes by which islets are formed and maintained. We find moves on β-cell graphs that leave the same significant aspects of cell arrangements unchanged. These moves turn out to be severely restricted, and suggest that β cells may prefer to move from larger clusters but can move to a cluster of any size, possibly to maximize their exposure to blood vessels.
Collapse
Affiliation(s)
- Deborah A. Striegel
- Laboratory of Biological Modeling, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Manami Hara
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Vipul Periwal
- Laboratory of Biological Modeling, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
78
|
Meigh L, Cook D, Zhang J, Dale N. Rational design of new NO and redox sensitivity into connexin26 hemichannels. Open Biol 2015; 5:140208. [PMID: 25673329 PMCID: PMC4345282 DOI: 10.1098/rsob.140208] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
CO2 directly opens hemichannels of connexin26 (Cx26) by carbamylating K125, thereby allowing salt bridge formation with R104 of the neighbouring subunit in the connexin hexamer. The formation of the inter-subunit carbamate bridges within the hexameric hemichannel traps it in the open state. Here, we use insights derived from this model to test whether the range of agonists capable of opening Cx26 can be extended by promoting the formation of analogous inter-subunit bridges via different mechanisms. The mutation K125C gives potential for nitrosylation on Cys125 and formation of an SNO bridge to R104 of the neighbouring subunit. Unlike wild-type Cx26 hemichannels, which are insensitive to NO and NO2−, hemichannels comprising Cx26K125C can be opened by NO2− and NO donors. However, NO2− was unable to modulate the doubly mutated (K125C, R104A) hemichannels, indicating that an inter-subunit bridge between C125 and R104 is required for the opening action of NO2−. In a further test, we introduced two mutations into Cx26, K125C and R104C, to allow disulfide bridge formation across the inter-subunit boundary. These doubly mutated hemichannels open in response to changes in intracellular redox potential.
Collapse
Affiliation(s)
- Louise Meigh
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Daniel Cook
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Jie Zhang
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Nicholas Dale
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| |
Collapse
|
79
|
Meens MJ, Alonso F, Le Gal L, Kwak BR, Haefliger JA. Endothelial Connexin37 and Connexin40 participate in basal but not agonist-induced NO release. Cell Commun Signal 2015; 13:34. [PMID: 26198171 PMCID: PMC4510910 DOI: 10.1186/s12964-015-0110-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 07/03/2015] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Connexin37 (Cx37) and Cx40 are crucial for endothelial cell-cell communication and homeostasis. Both connexins interact with endothelial nitric oxide synthase (eNOS). The exact contribution of these interactions to the regulation of vascular tone is unknown. RESULTS Cx37 and Cx40 were expressed in close proximity to eNOS at cell-cell interfaces of mouse aortic endothelial cells. Absence of Cx37 did not affect expression of Cx40 and a 50 % reduction of Cx40 in Cx40(+/-) aortas did not affect the expression of Cx37. However, absence of Cx40 was associated with reduced expression of Cx37. Basal NO release and the sensitivity for ACh were decreased in Cx37(-/-) and Cx40(-/-) aortas but not in Cx40(+/-) aortas. Moreover, ACh-induced release of constricting cyclooxygenase products was present in WT, Cx40(-/-) and Cx40(+/-) aortas but not in Cx37(-/-) aortas. Finally, agonist-induced NO-dependent relaxations and the sensitivity for exogenous NO were not affected by genotype. CONCLUSIONS Cx37 is more markedly involved in basal NO release, release of cyclooxygenase products and the regulation of the sensitivity for ACh as compared to Cx40.
Collapse
Affiliation(s)
- Merlijn J Meens
- Department of Pathology and Immunology, University of Geneva, 6th floor, 1 Rue Michel-Servet, 1211, Geneva, Switzerland.
- Department of Medical Specialties - Cardiology, University of Geneva, Geneva, Switzerland.
| | - Florian Alonso
- Department of Medicine, University Hospital, CHUV, Lausanne, Switzerland
| | - Loïc Le Gal
- Department of Medicine, University Hospital, CHUV, Lausanne, Switzerland
| | - Brenda R Kwak
- Department of Pathology and Immunology, University of Geneva, 6th floor, 1 Rue Michel-Servet, 1211, Geneva, Switzerland
- Department of Medical Specialties - Cardiology, University of Geneva, Geneva, Switzerland
| | | |
Collapse
|
80
|
Preda MB, Rosca AM, Tutuianu R, Burlacu A. Pre-stimulation with FGF-2 increases in vitro functional coupling of mesenchymal stem cells with cardiac cells. Biochem Biophys Res Commun 2015; 464:667-73. [PMID: 26187662 DOI: 10.1016/j.bbrc.2015.07.055] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 07/09/2015] [Indexed: 11/27/2022]
Abstract
The functional coupling of transplanted cells with host myocardial cells is a significant challenge in mesenchymal stem cell (MSC) cardiomyoplasty being related to cell survival and therapeutic outcomes. Priming of MSCs with growth factors has been reported to improve their therapeutic efficacy through gap junction-mediated mechanisms. However, the expression pattern of Connexin43 (Cx43) in growth factor-stimulated MSC was not previously addressed. In this study we investigated how the pre-treatment with growth factors modulates MSC ability to integrate into the host tissue after transplantation, with particular focus on the expression of Cx43 and its cellular distribution. Our results showed that stimulation of MSCs with IGF-1, FGF-2, but not TGFβ, increased the level of Cx43 at both mRNA and protein levels. IGF-1 stimulation resulted in a shift of the fibroblast morphology into an epithelial morphology in several well-defined areas of stimulated cells. Confocal microscopy examination revealed that the increase of Cx43 was restricted to the epithelial-like cells and did not occur in other cells. In variance, FGF-2 induced a rod-shape morphology of every single cell, which achieved an extremely low cell index. FGF-2 stimulation also induced a time-dependent increase in Cx43, with a regular distribution pattern in all cells. Dye transfer assay coupled with confocal microscopy and flow cytometry analysis demonstrated functional in vitro cell coupling between FGF-2-stimulated MSCs as well as between FGF-2-stimulated cells and H9c2 cardiomyoblasts, a scenery that mimick MSC transplantation into the myocardium. We conclude that the stimulation of MSCs with FGF-2 prior to transplantation may facilitate their access among the myocardial cells and increase the functional coupling between transplanted and host cells.
Collapse
Affiliation(s)
- Mihai Bogdan Preda
- Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Ana-Maria Rosca
- Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Raluca Tutuianu
- Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Alexandrina Burlacu
- Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania.
| |
Collapse
|
81
|
Hodson DJ, Legros C, Desarménien MG, Guérineau NC. Roles of connexins and pannexins in (neuro)endocrine physiology. Cell Mol Life Sci 2015; 72:2911-28. [PMID: 26084873 DOI: 10.1007/s00018-015-1967-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 06/11/2015] [Indexed: 12/12/2022]
Abstract
To ensure appropriate secretion in response to demand, (neuro)endocrine tissues liberate massive quantities of hormones, which act to coordinate and synchronize biological signals in distant secretory and nonsecretory cell populations. Intercellular communication plays a central role in this control. With regard to molecular identity, junctional cell-cell communication is supported by connexin-based gap junctions. In addition, connexin hemichannels, the structural precursors of gap junctions, as well as pannexin channels have recently emerged as possible modulators of the secretory process. This review focuses on the expression of connexins and pannexins in various (neuro)endocrine tissues, including the adrenal cortex and medulla, the anterior pituitary, the endocrine hypothalamus and the pineal, thyroid and parathyroid glands. Upon a physiological or pathological stimulus, junctional intercellular coupling can be acutely modulated or persistently remodeled, thus offering multiple regulatory possibilities. The functional roles of gap junction-mediated intercellular communication in endocrine physiology as well as the involvement of connexin/pannexin-related hemichannels are also discussed.
Collapse
Affiliation(s)
- David J Hodson
- Section of Cell Biology and Functional Genomics, Department of Medicine, Imperial College London, London, W12 0NN, UK
| | | | | | | |
Collapse
|
82
|
Lebreton F, Pirog A, Belouah I, Bosco D, Berney T, Meda P, Bornat Y, Catargi B, Renaud S, Raoux M, Lang J. Slow potentials encode intercellular coupling and insulin demand in pancreatic beta cells. Diabetologia 2015; 58:1291-9. [PMID: 25788295 DOI: 10.1007/s00125-015-3558-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 02/23/2015] [Indexed: 11/24/2022]
Abstract
AIMS/HYPOTHESIS Ion fluxes constitute a major integrative signal in beta cells that leads to insulin secretion and regulation of gene expression. Understanding these electrical signals is important for deciphering the endogenous algorithms used by islets to attain homeostasis and for the design of new sensors for monitoring beta cell function. METHODS Mouse and human islets were cultured on multielectrode arrays (MEAs) for 3-13 days. Extracellular electrical activities received on each electrode were continuously amplified and recorded for offline characterisation. RESULTS Differential band-pass filtering of MEA recordings of mouse islets showed two extracellular voltage waveforms: action potentials (lasting 40-60 ms) and very robust slow potentials (SPs, lasting 800-1,500 ms), the latter of which have not been described previously. The frequency of SPs directly correlated with glucose concentration, peaked at 10 mmol/l glucose and was further augmented by picomolar concentrations of glucagon-like peptide-1. SPs required the closure of ATP-dependent potassium channels as they were induced by glucose or glibenclamide but were not elicited by KCl-induced depolarisation. Pharmacological tools and the use of beta cell specific knockout mice showed that SPs reflected cell coupling via connexin 36. Moreover, increasing and decreasing glucose ramps showed hysteresis with reduced glucose sensitivity during the decreasing phase. SPs were also observed in human islets and could be continuously recorded over 24 h. CONCLUSIONS/INTERPRETATION This novel electrical signature reflects the syncytial function of the islets and is specific to beta cells. Moreover, the observed hysteresis provides evidence for an endogenous algorithm naturally present in islets to protect against hypoglycaemia.
Collapse
Affiliation(s)
- Fanny Lebreton
- CNRS UMR 5248, Chimie et Biologie des Membranes et Nano-objets, Université de Bordeaux, Batiment B14, Allée Geoffroy St Hilaire, CS90063, 33615, Pessac, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Kim SM, Lee EJ, Jung HS, Han N, Kim YJ, Kim TK, Kim TN, Kwon MJ, Lee SH, Park JH, Rhee BD, Kim MK. Co-Culture of α TC-6 Cells and β TC-1 Cells: Morphology and Function. Endocrinol Metab (Seoul) 2015; 30:92-7. [PMID: 25325280 PMCID: PMC4384678 DOI: 10.3803/enm.2015.30.1.92] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 07/14/2014] [Accepted: 08/05/2014] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND In vitro experiments using only β-cell lines instead of islets are limited because pancreatic islets are composed of four different types of endocrine cells. Several recent studies have focused on cellular interactions among these cell types, especially α- and β-cells. Because islet isolation needs time and experience, we tested a simple co-culture system with α- and β-cells. Their morphology and function were assessed by comparison to each single cell culture and pancreatic islets. METHODS α TC-6 cells and β TC-1 cells were maintained in Dulbecco's Minimal Essential Medium containing 5 mM glucose and 10% fetal bovine serum. Cells were mixed at a 1:1 ratio (5×10⁵) in 6-well plates and cultured for 24, 48, and 72 hours. After culture, cells were used for insulin and glucagon immunoassays and tested for glucose-stimulated insulin secretion (GSIS). RESULTS α TC-6 and β TC-1 cells became condensed by 24 hours and were more strongly compacted after 48 hours. β TC-1 cells showed both β-β and β-α cell contacts. GSIS increased with increasing glucose concentration in co-cultured cells, which showed lower secreted insulin levels than β TC-1 cells alone. The increase in the secreted insulin/insulin content ratio was significantly lower for co-cultured cells than for β-cells alone (P=0.04). Compared to islets, the α-/β-cell co-culture showed a higher ratio of GSIS to insulin content, but the difference was not statistically significant (P=0.09). CONCLUSION α TC-6 and β TC-1 cells in the co-culture system showed cell-to-cell contacts and a similar stimulated insulin secretion pattern to islets. The co-culture system may be used to better mimic pancreatic islets in in vitro assessments.
Collapse
Affiliation(s)
- Sung Man Kim
- Department of Internal Medicine, Inje University College of Medicine, Busan, Korea
| | - Eun Ju Lee
- Department of Internal Medicine, Inje University College of Medicine, Busan, Korea
| | - Hye Sook Jung
- Moelcular Therapy Lab, Paik Memorial Institute for Clinical Research, Inje University, Busan, Korea
| | - Na Han
- Department of Internal Medicine, Inje University College of Medicine, Busan, Korea
| | - You Jeong Kim
- Department of Internal Medicine, Inje University College of Medicine, Busan, Korea
| | - Tae Kyoon Kim
- Department of Internal Medicine, Inje University College of Medicine, Busan, Korea
| | - Tae Nyun Kim
- Department of Internal Medicine, Inje University College of Medicine, Busan, Korea
| | - Min Jeong Kwon
- Department of Internal Medicine, Inje University College of Medicine, Busan, Korea
| | - Soon Hee Lee
- Department of Internal Medicine, Inje University College of Medicine, Busan, Korea
| | - Jeong Hyun Park
- Department of Internal Medicine, Inje University College of Medicine, Busan, Korea.; Moelcular Therapy Lab, Paik Memorial Institute for Clinical Research, Inje University, Busan, Korea
| | - Byoung Doo Rhee
- Department of Internal Medicine, Inje University College of Medicine, Busan, Korea
| | - Mi Kyung Kim
- Department of Internal Medicine, Inje University College of Medicine, Busan, Korea.; Moelcular Therapy Lab, Paik Memorial Institute for Clinical Research, Inje University, Busan, Korea.
| |
Collapse
|
84
|
Katoch P, Mitra S, Ray A, Kelsey L, Roberts BJ, Wahl JK, Johnson KR, Mehta PP. The carboxyl tail of connexin32 regulates gap junction assembly in human prostate and pancreatic cancer cells. J Biol Chem 2015; 290:4647-4662. [PMID: 25548281 PMCID: PMC4335205 DOI: 10.1074/jbc.m114.586057] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 12/23/2014] [Indexed: 12/20/2022] Open
Abstract
Connexins, the constituent proteins of gap junctions, are transmembrane proteins. A connexin (Cx) traverses the membrane four times and has one intracellular and two extracellular loops with the amino and carboxyl termini facing the cytoplasm. The transmembrane and the extracellular loop domains are highly conserved among different Cxs, whereas the carboxyl termini, often called the cytoplasmic tails, are highly divergent. We have explored the role of the cytoplasmic tail of Cx32, a Cx expressed in polarized and differentiated cells, in regulating gap junction assembly. Our results demonstrate that compared with the full-length Cx32, the cytoplasmic tail-deleted Cx32 is assembled into small gap junctions in human pancreatic and prostatic cancer cells. Our results further document that the expression of the full-length Cx32 in cells, which express the tail-deleted Cx32, increases the size of gap junctions, whereas the expression of the tail-deleted Cx32 in cells, which express the full-length Cx32, has the opposite effect. Moreover, we show that the tail is required for the clustering of cell-cell channels and that in cells expressing the tail-deleted Cx32, the expression of cell surface-targeted cytoplasmic tail alone is sufficient to enhance the size of gap junctions. Our live-cell imaging data further demonstrate that gap junctions formed of the tail-deleted Cx32 are highly mobile compared with those formed of full-length Cx32. Our results suggest that the cytoplasmic tail of Cx32 is not required to initiate the assembly of gap junctions but for their subsequent growth and stability. Our findings suggest that the cytoplasmic tail of Cx32 may be involved in regulating the permeability of gap junctions by regulating their size.
Collapse
Affiliation(s)
- Parul Katoch
- From the Department of Biochemistry and Molecular Biology, Department of Oral Biology, Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Shalini Mitra
- From the Department of Biochemistry and Molecular Biology, Department of Oral Biology, Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Anuttoma Ray
- From the Department of Biochemistry and Molecular Biology, Department of Oral Biology, Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Linda Kelsey
- From the Department of Biochemistry and Molecular Biology, Department of Oral Biology, Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Brett J Roberts
- From the Department of Biochemistry and Molecular Biology, Department of Oral Biology, Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - James K Wahl
- From the Department of Biochemistry and Molecular Biology, Department of Oral Biology, Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Keith R Johnson
- From the Department of Biochemistry and Molecular Biology, Department of Oral Biology, Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Parmender P Mehta
- From the Department of Biochemistry and Molecular Biology, Department of Oral Biology, Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198.
| |
Collapse
|
85
|
Kaneko Y, Tachikawa M, Akaogi R, Fujimoto K, Ishibashi M, Uchida Y, Couraud PO, Ohtsuki S, Hosoya KI, Terasaki T. Contribution of pannexin 1 and connexin 43 hemichannels to extracellular calcium-dependent transport dynamics in human blood-brain barrier endothelial cells. J Pharmacol Exp Ther 2015; 353:192-200. [PMID: 25670633 DOI: 10.1124/jpet.114.220210] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Dysregulation of blood-brain barrier (BBB) transport function is thought to exacerbate neuronal damage in acute ischemic stroke. The purpose of this study was to clarify the characteristics of pannexin (Px) and/or connexin (Cx) hemichannel(s)-mediated transport of organic anions and cations in human BBB endothelial cell line hCMEC/D3 and to identify inhibitors of hemichannel opening in hCMEC/D3 cells in the absence of extracellular Ca(2+), a condition mimicking acute ischemic stroke. In the absence of extracellular Ca(2+), the cells showed increased uptake and efflux transport of organic ionic fluorescent dyes. Classic hemichannel inhibitors markedly inhibited the enhanced uptake and efflux. Quantitative targeted absolute proteomics confirmed Px1 and Cx43 protein expression in plasma membrane of hCMEC/D3 cells. Knockdown of Px1 and Cx43 with the small interfering RNAs significantly inhibited the enhanced uptake and efflux of organic anionic and cationic fluorescent dyes. Clinically used cilnidipine and progesterone, which have neuroprotective effects in animal ischemia models, were identified as inhibitors of hemichannel opening. These findings suggest that altered transport dynamics at the human BBB in the absence of extracellular Ca(2+) is at least partly attributable to opening of Px1 and Cx43 hemichannels. Therefore, we speculate that Px1 and Cx43 may be potential drug targets to ameliorate BBB transport dysregulation during acute ischemia.
Collapse
Affiliation(s)
- Yosuke Kaneko
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (Y.K., M.T., R.A., Y.U., T.T.); Department of Applied Chemistry and Biochemistry, Faculty of Engineering, Kyushu Sangyo University, Fukuoka, Japan (K.F., M.I.); INSERM, U1016, Institut Cochin and CNRS, UMR8104, and Université Paris Descartes, Paris, France (P.-O.C.); Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan (S.O.); and Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (K.H.)
| | - Masanori Tachikawa
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (Y.K., M.T., R.A., Y.U., T.T.); Department of Applied Chemistry and Biochemistry, Faculty of Engineering, Kyushu Sangyo University, Fukuoka, Japan (K.F., M.I.); INSERM, U1016, Institut Cochin and CNRS, UMR8104, and Université Paris Descartes, Paris, France (P.-O.C.); Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan (S.O.); and Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (K.H.)
| | - Ryo Akaogi
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (Y.K., M.T., R.A., Y.U., T.T.); Department of Applied Chemistry and Biochemistry, Faculty of Engineering, Kyushu Sangyo University, Fukuoka, Japan (K.F., M.I.); INSERM, U1016, Institut Cochin and CNRS, UMR8104, and Université Paris Descartes, Paris, France (P.-O.C.); Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan (S.O.); and Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (K.H.)
| | - Kazuhisa Fujimoto
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (Y.K., M.T., R.A., Y.U., T.T.); Department of Applied Chemistry and Biochemistry, Faculty of Engineering, Kyushu Sangyo University, Fukuoka, Japan (K.F., M.I.); INSERM, U1016, Institut Cochin and CNRS, UMR8104, and Université Paris Descartes, Paris, France (P.-O.C.); Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan (S.O.); and Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (K.H.)
| | - Megumi Ishibashi
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (Y.K., M.T., R.A., Y.U., T.T.); Department of Applied Chemistry and Biochemistry, Faculty of Engineering, Kyushu Sangyo University, Fukuoka, Japan (K.F., M.I.); INSERM, U1016, Institut Cochin and CNRS, UMR8104, and Université Paris Descartes, Paris, France (P.-O.C.); Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan (S.O.); and Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (K.H.)
| | - Yasuo Uchida
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (Y.K., M.T., R.A., Y.U., T.T.); Department of Applied Chemistry and Biochemistry, Faculty of Engineering, Kyushu Sangyo University, Fukuoka, Japan (K.F., M.I.); INSERM, U1016, Institut Cochin and CNRS, UMR8104, and Université Paris Descartes, Paris, France (P.-O.C.); Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan (S.O.); and Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (K.H.)
| | - Pierre-Olivier Couraud
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (Y.K., M.T., R.A., Y.U., T.T.); Department of Applied Chemistry and Biochemistry, Faculty of Engineering, Kyushu Sangyo University, Fukuoka, Japan (K.F., M.I.); INSERM, U1016, Institut Cochin and CNRS, UMR8104, and Université Paris Descartes, Paris, France (P.-O.C.); Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan (S.O.); and Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (K.H.)
| | - Sumio Ohtsuki
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (Y.K., M.T., R.A., Y.U., T.T.); Department of Applied Chemistry and Biochemistry, Faculty of Engineering, Kyushu Sangyo University, Fukuoka, Japan (K.F., M.I.); INSERM, U1016, Institut Cochin and CNRS, UMR8104, and Université Paris Descartes, Paris, France (P.-O.C.); Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan (S.O.); and Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (K.H.)
| | - Ken-ichi Hosoya
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (Y.K., M.T., R.A., Y.U., T.T.); Department of Applied Chemistry and Biochemistry, Faculty of Engineering, Kyushu Sangyo University, Fukuoka, Japan (K.F., M.I.); INSERM, U1016, Institut Cochin and CNRS, UMR8104, and Université Paris Descartes, Paris, France (P.-O.C.); Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan (S.O.); and Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (K.H.)
| | - Tetsuya Terasaki
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan (Y.K., M.T., R.A., Y.U., T.T.); Department of Applied Chemistry and Biochemistry, Faculty of Engineering, Kyushu Sangyo University, Fukuoka, Japan (K.F., M.I.); INSERM, U1016, Institut Cochin and CNRS, UMR8104, and Université Paris Descartes, Paris, France (P.-O.C.); Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan (S.O.); and Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan (K.H.)
| |
Collapse
|
86
|
Hills CE, Price GW, Squires PE. Mind the gap: connexins and cell-cell communication in the diabetic kidney. Diabetologia 2015; 58:233-41. [PMID: 25358446 DOI: 10.1007/s00125-014-3427-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 09/25/2014] [Indexed: 10/24/2022]
Abstract
Connexins, assembled as a hexameric connexon, form a transmembrane hemichannel that provides a conduit for paracrine signalling of small molecules and ions to regulate the activity and function of adjacent cells. When hemichannels align and associate with similar channels on opposing cells, they form a continuous aqueous pore or gap junction, allowing the direct transmission of metabolic and electrical signals between coupled cells. Regulation of gap junction synthesis and channel activity is critical for cell function, and a number of diseases can be attributed to changes in the expression/function of these important proteins. Diabetic nephropathy is associated with several complex metabolic and inflammatory responses characterised by defects at the molecular, cellular and tissue level. In both type 1 and type 2 diabetes, glycaemic injury of the kidney is the leading cause of end-stage renal failure, a consequence of multiple aetiologies, including increased deposition of extracellular matrix, glomerular hyperfiltration, albuminuria and tubulointerstitial fibrosis. In diabetic nephropathy, loss of connexin mediated cell-cell communication within the nephron may represent an early sign of disease; however, our current knowledge of the role of connexins in the diabetic kidney is sparse. This review highlights recent evidence demonstrating that maintenance of connexin-mediated cell-cell communication could benefit region-specific renal function in diabetic nephropathy and suggests that these proteins should be viewed as a tantalising novel target for therapeutic intervention.
Collapse
Affiliation(s)
- Claire E Hills
- School of Life Sciences, University of Lincoln, Brayford Pool, Lincoln, LN6 7TS, UK,
| | | | | |
Collapse
|
87
|
Rutter GA, Hodson DJ. Beta cell connectivity in pancreatic islets: a type 2 diabetes target? Cell Mol Life Sci 2015; 72:453-467. [PMID: 25323131 PMCID: PMC11113448 DOI: 10.1007/s00018-014-1755-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 09/30/2014] [Accepted: 10/01/2014] [Indexed: 12/12/2022]
Abstract
Beta cell connectivity describes the phenomenon whereby the islet context improves insulin secretion by providing a three-dimensional platform for intercellular signaling processes. Thus, the precise flow of information through homotypically interconnected beta cells leads to the large-scale organization of hormone release activities, influencing cell responses to glucose and other secretagogues. Although a phenomenon whose importance has arguably been underappreciated in islet biology until recently, a growing number of studies suggest that such cell-cell communication is a fundamental property of this micro-organ. Hence, connectivity may plausibly be targeted by both environmental and genetic factors in type 2 diabetes mellitus (T2DM) to perturb normal beta cell function and insulin release. Here, we review the mechanisms that contribute to beta cell connectivity, discuss how these may fail during T2DM, and examine approaches to restore insulin secretion by boosting cell communication.
Collapse
Affiliation(s)
- Guy A Rutter
- Section of Cell Biology, Department of Medicine, Imperial College London, Imperial Centre for Translational and Experimental Medicine, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK.
| | - David J Hodson
- Section of Cell Biology, Department of Medicine, Imperial College London, Imperial Centre for Translational and Experimental Medicine, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
88
|
Calkins TL, Woods-Acevedo MA, Hildebrandt O, Piermarini PM. The molecular and immunochemical expression of innexins in the yellow fever mosquito, Aedes aegypti: insights into putative life stage- and tissue-specific functions of gap junctions. Comp Biochem Physiol B Biochem Mol Biol 2015; 183:11-21. [PMID: 25585357 DOI: 10.1016/j.cbpb.2014.11.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 11/17/2014] [Accepted: 11/21/2014] [Indexed: 11/28/2022]
Abstract
Gap junctions (GJ) mediate direct intercellular communication by forming channels through which certain small molecules and/or ions can pass. Connexins, the proteins that form vertebrate GJ, are well studied and known to contribute to neuronal, muscular and epithelial physiology. Innexins, the GJ proteins of insects, have only recently received much investigative attention and many of their physiological roles remain to be determined. Here we characterize the molecular expression of six innexin (Inx) genes in the yellow fever mosquito Aedes aegypti (AeInx1, AeInx2, AeInx3, AeInx4, AeInx7, and AeInx8) and the immunochemical expression of one innexin protein, AeInx3, in the alimentary canal. We detected the expression of no less than four innexin genes in each mosquito life stage (larva, pupa, adult) and tissue/body region from adult males and females (midgut, Malpighian tubules, hindgut, head, carcass, gonads), suggesting a remarkable potential molecular diversity of GJ in mosquitoes. Moreover, the expression patterns of some innexins were life stage and/or tissue specific, suggestive of potential functional specializations. Cloning of the four full-length cDNAs expressed in the Malpighian tubules of adult females (AeInx1, AeInx2, AeInx3, and AeInx7) revealed evidence for 1) alternative splicing of AeInx1 and AeInx3 transcripts, and 2) putative N-glycosylation of AeInx3 and AeInx7. Finally, immunohistochemistry of AeInx3 in the alimentary canal of larval and adult female mosquitoes confirmed localization of this innexin to the intercellular regions of Malpighian tubule and hindgut epithelial cells, suggesting that it is an important component of GJ in these tissues.
Collapse
Affiliation(s)
- Travis L Calkins
- Department of Entomology, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, United States
| | - Mikal A Woods-Acevedo
- Department of Entomology, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, United States
| | - Oliver Hildebrandt
- Department of Entomology, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, United States
| | - Peter M Piermarini
- Department of Entomology, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, United States.
| |
Collapse
|
89
|
Liu X, Yan F, Yao H, Chang M, Qin J, Li Y, Wang Y, Pei X. Involvement of RhoA/ROCK in insulin secretion of pancreatic β-cells in 3D culture. Cell Tissue Res 2014; 358:359-69. [PMID: 25129107 DOI: 10.1007/s00441-014-1961-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 07/03/2014] [Indexed: 01/07/2023]
Abstract
Cell-cell contacts and interactions between pancreatic β-cells and/or other cell populations within islets are essential for cell survival, insulin secretion, and functional synchronization. Three-dimensional (3D) culture systems supply the ideal microenvironment for islet-like cluster formation and functional maintenance. However, the underlying mechanisms remain unclear. In this study, mouse insulinoma 6 (MIN6) cells were cultured in a rotating 3D culture system to form islet-like aggregates. Glucose-stimulated insulin secretion (GSIS) and the RhoA/ROCK pathway were investigated. In the 3D-cultured MIN6 cells, more endocrine-specific genes were up-regulated, and GSIS was increased to a greater extent than in cells grown in monolayers. RhoA/ROCK inactivation led to F-actin remodeling in the MIN6 cell aggregates and greater insulin exocytosis. The gap junction protein, connexin 36 (Cx36), was up-regulated in MIN6 cell aggregates and RhoA/ROCK-inactivated monolayer cells. GSIS dramatically decreased when Cx36 was knocked down by short interfering RNA and could not be reversed by RhoA/ROCK inactivation. Thus, the RhoA/ROCK signaling pathway is involved in insulin release through the up-regulation of Cx36 expression in 3D-cultured MIN6 cells.
Collapse
Affiliation(s)
- Xiaofang Liu
- Stem Cell and Regenerative Medicine Laboratory, Beijing Institute of Transfusion Medicine, Beijing, 100850, China
| | | | | | | | | | | | | | | |
Collapse
|
90
|
Kelsey L, Katoch P, Ray A, Mitra S, Chakraborty S, Lin MF, Mehta PP. Vitamin D3 regulates the formation and degradation of gap junctions in androgen-responsive human prostate cancer cells. PLoS One 2014; 9:e106437. [PMID: 25188420 PMCID: PMC4154685 DOI: 10.1371/journal.pone.0106437] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 08/06/2014] [Indexed: 11/19/2022] Open
Abstract
1α-25(OH)2 vitamin D3 (1-25D), an active hormonal form of Vitamin D3, is a well-known chemopreventive and pro-differentiating agent. It has been shown to inhibit the growth of several prostate cancer cell lines. Gap junctions, formed of proteins called connexins (Cx), are ensembles of cell-cell channels, which permit the exchange of small growth regulatory molecules between adjoining cells. Cell-cell communication mediated by gap junctional channels is an important homeostatic control mechanism for regulating cell growth and differentiation. We have investigated the effect of 1-25D on the formation and degradation of gap junctions in an androgen-responsive prostate cancer cell line, LNCaP, which expresses retrovirally-introduced Cx32. Connexin32 is expressed by the luminal and well-differentiated cells of normal prostate and prostate tumors. Our results document that 1-25D enhances the expression of Cx32 and its subsequent assembly into gap junctions. Our results further show that 1-25D prevents androgen-regulated degradation of Cx32, post-translationally, independent of androgen receptor (AR)-mediated signaling. Finally, our findings document that formation of gap junctions sensitizes Cx32-expressing LNCaP cells to the growth inhibitory effects of 1-25D and alters their morphology. These findings suggest that the growth-inhibitory effects of 1-25D in LNCaP cells may be related to its ability to modulate the assembly of Cx32 into gap junctions.
Collapse
Affiliation(s)
- Linda Kelsey
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Parul Katoch
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Anuttoma Ray
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Shalini Mitra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Souvik Chakraborty
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Ming-Fong Lin
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Parmender P. Mehta
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| |
Collapse
|
91
|
Benninger RKP, Piston DW. Cellular communication and heterogeneity in pancreatic islet insulin secretion dynamics. Trends Endocrinol Metab 2014; 25:399-406. [PMID: 24679927 PMCID: PMC4112137 DOI: 10.1016/j.tem.2014.02.005] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 02/21/2014] [Accepted: 02/25/2014] [Indexed: 11/20/2022]
Abstract
Coordinated pulses of electrical activity and insulin secretion are a hallmark of the islet of Langerhans. These coordinated behaviors are lost when β cells are dissociated, which also leads to increased insulin secretion at low glucose levels. Islets without gap junctions exhibit asynchronous electrical activity similar to dispersed cells, but their secretion at low glucose levels is still clamped off, putatively by a juxtacrine mechanism. Mice lacking β cell gap junctions have near-normal average insulin levels, but are glucose intolerant due to reduced first-phase and pulsatile insulin secretion, illustrating the importance of temporal dynamics. Here, we review the quantitative data on islet synchronization and the current mathematical models that have been developed to explain these behaviors and generate greater understanding of the underlying mechanisms.
Collapse
Affiliation(s)
- Richard K P Benninger
- Department of Bioengineering and Barbara Davis Center, University of Colorado Anschutz Medical campus, Aurora, CO, USA.
| | - David W Piston
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
92
|
Spéder P, Brand AH. Gap junction proteins in the blood-brain barrier control nutrient-dependent reactivation of Drosophila neural stem cells. Dev Cell 2014; 30:309-21. [PMID: 25065772 PMCID: PMC4139190 DOI: 10.1016/j.devcel.2014.05.021] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 04/15/2014] [Accepted: 05/24/2014] [Indexed: 01/27/2023]
Abstract
Neural stem cells in the adult brain exist primarily in a quiescent state but are reactivated in response to changing physiological conditions. How do stem cells sense and respond to metabolic changes? In the Drosophila CNS, quiescent neural stem cells are reactivated synchronously in response to a nutritional stimulus. Feeding triggers insulin production by blood-brain barrier glial cells, activating the insulin/insulin-like growth factor pathway in underlying neural stem cells and stimulating their growth and proliferation. Here we show that gap junctions in the blood-brain barrier glia mediate the influence of metabolic changes on stem cell behavior, enabling glia to respond to nutritional signals and reactivate quiescent stem cells. We propose that gap junctions in the blood-brain barrier are required to translate metabolic signals into synchronized calcium pulses and insulin secretion. Blood-brain barrier gap junctions are required for neural stem cell reactivation Gap junctions control both insulin transcription and secretion Calcium oscillations in the blood-brain barrier depend on gap junctions and nutrition Blood-brain barrier membrane polarization links calcium to insulin secretion
Collapse
Affiliation(s)
- Pauline Spéder
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Andrea H Brand
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK.
| |
Collapse
|
93
|
Le Gal L, Alonso F, Wagner C, Germain S, Nardelli Haefliger D, Meda P, Haefliger JA. Restoration of connexin 40 (Cx40) in Renin-producing cells reduces the hypertension of Cx40 null mice. Hypertension 2014; 63:1198-204. [PMID: 24614215 DOI: 10.1161/hypertensionaha.113.02976] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Connexin 40 (Cx40) is expressed by the renin-producing cells (RSCs) of the kidneys and the endothelial cells of blood vessels. Cx40 null mice (Cx40(-/-)) feature a much increased renin synthesis and secretion, which results in chronic hypertension, and also display an altered endothelium-dependent relaxation of the aorta because of reduced eNOS levels and nitric oxide production. To discriminate the effect of Cx40 in renin secretion and vascular signaling, we targeted Cx40 to either the RSCs or the endothelial cells of Cx40 null mice. When compared with Cx40(-/-) controls, the animals expressing Cx40 in RSCs were less hypertensive and featured reduced renin levels, still numerous RSCs outside the wall of the afferent arterioles. In contrast, mice expressing Cx40 in the endothelial cells were as hypertensive as Cx40(-/-) mice, in spite of control levels of Cx37 and eNOS. Our data show that blood pressure is improved by restoration of Cx40 expression in RSCs but not in endothelial cells, stressing the prominent role of renin in the mouse hypertension linked to loss of Cx40.
Collapse
Affiliation(s)
- Loïc Le Gal
- Department of Medicine, Laboratory of Experimental Medicine, c/o Department of Physiology, Bugnon 7a, 1005 Lausanne, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
94
|
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
|
95
|
Farnsworth NL, Benninger RKP. New insights into the role of connexins in pancreatic islet function and diabetes. FEBS Lett 2014; 588:1278-87. [PMID: 24583073 DOI: 10.1016/j.febslet.2014.02.035] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Revised: 02/13/2014] [Accepted: 02/19/2014] [Indexed: 12/22/2022]
Abstract
Multi-cellular systems require complex signaling mechanisms for proper tissue function, to mediate signaling between cells in close proximity and at distances. This holds true for the islets of Langerhans, which are multicellular micro-organs located in the pancreas responsible for glycemic control, through secretion of insulin and other hormones. Coupling of electrical and metabolic signaling between islet β-cells is required for proper insulin secretion and effective glycemic control. β-cell specific coupling is established through gap junctions composed of connexin36, which results in coordinated insulin release across the islet. Islet connexins have been implicated in both Type-1 and Type-2 diabetes; however a clear link remains to be determined. The goal of this review is to discuss recent discoveries regarding the role of connexins in regulating insulin secretion, the regulation of connexins within the islet, and recent studies which support a role for connexins in diabetes. Further studies which investigate the regulation of connexins in the islet and their role in diabetes may lead to novel diabetes therapies which regulate islet function and β-cell survival through modulation of gap junction coupling.
Collapse
Affiliation(s)
- Nikki L Farnsworth
- Barbara Davis center for childhood diabetes, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Richard K P Benninger
- Barbara Davis center for childhood diabetes, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, United States; Department of Bioengineering, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, United States.
| |
Collapse
|
96
|
Review: Novel insights into the regulation of vascular tone by sphingosine 1-phosphate. Placenta 2014; 35 Suppl:S86-92. [DOI: 10.1016/j.placenta.2013.12.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 12/10/2013] [Accepted: 12/11/2013] [Indexed: 11/17/2022]
|
97
|
Rafacho A. Effects of glucocorticoids and exercise on pancreatic β-cell function and diabetes development: comments on Beaudry and Riddel. Diabetes Metab Res Rev 2014; 30:120-1. [PMID: 24500990 DOI: 10.1002/dmrr.2473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 09/10/2013] [Indexed: 11/11/2022]
Affiliation(s)
- Alex Rafacho
- Department of Physiological Sciences, Centre of Biological Sciences, Federal University of Santa Catarina (UFSC), 88040-900 Florianópolis, SC, Brazil
| |
Collapse
|
98
|
Meens MJ, Sabine A, Petrova TV, Kwak BR. Connexins in lymphatic vessel physiology and disease. FEBS Lett 2014; 588:1271-7. [PMID: 24457200 DOI: 10.1016/j.febslet.2014.01.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 01/13/2014] [Accepted: 01/14/2014] [Indexed: 12/26/2022]
Abstract
Connexins are transmembrane proteins that form gap junction- and hemi-channels. Once inserted into the membrane, hemi-channels (connexons) allow for diffusion of ions and small molecules (<1 kDa) between the extracellular space and the cytosol. Gap junction channels allow diffusion of similar molecules between the cytoplasms of adjacent cells. The expression and function of connexins in blood vessels has been intensely studied in the last few decades. In contrast, only a few studies paid attention to lymphatic vessels; convincing in vivo data with respect to expression patterns of lymphatic connexins and their functional roles have only recently begun to emerge. Interestingly, mutations in connexin genes have been linked to diseases of lymphatic vasculature, most notably primary and secondary lymphedema. This review summarizes the available data regarding lymphatic connexins. More specifically it addresses (i) early studies aimed at presence of gap junction-like structures in lymphatic vessels, (ii) more recent studies focusing on lymphatic connexins using genetically engineered mice, and (iii) results of clinical studies that have reported lymphedema-linked mutations in connexin genes.
Collapse
Affiliation(s)
- Merlijn J Meens
- Department of Pathology and Immunology, University of Geneva, CH-1211 Geneva, Switzerland; Department of Internal Medicine - Cardiology, University of Geneva, CH-1211 Geneva, Switzerland
| | - Amélie Sabine
- Department of Oncology, University Hospital of Lausanne, 1066 Epalinges, Switzerland; Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland
| | - Tatiana V Petrova
- Department of Oncology, University Hospital of Lausanne, 1066 Epalinges, Switzerland; Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland; École Polytechnique Fédérale de Lausanne (EPFL), Swiss Institute for Experimental Cancer Research (ISREC), 1015 Lausanne, Switzerland
| | - Brenda R Kwak
- Department of Pathology and Immunology, University of Geneva, CH-1211 Geneva, Switzerland; Department of Internal Medicine - Cardiology, University of Geneva, CH-1211 Geneva, Switzerland.
| |
Collapse
|
99
|
Pérez-Armendariz EM. Connexin 36, a key element in pancreatic beta cell function. Neuropharmacology 2013; 75:557-66. [PMID: 23973309 DOI: 10.1016/j.neuropharm.2013.08.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 08/05/2013] [Accepted: 08/07/2013] [Indexed: 12/01/2022]
Abstract
The prevalence of diabetes at a global scale has markedly increased during the last three decades. Diabetes is a chronic disease that includes a group of metabolic disorders, in which high serum glucose levels is a common factor. Insulin is the only hormone that decreases serum glucose levels. Therefore, it is relevant to deepen our understanding of cell mechanisms that regulate insulin production and release. Insulin is produced in pancreatic islet beta cells. They are excitable cells and most of them are electrically coupled through gap junction channels. Connexin 36 (Cx36) has been identified at junctional membranes of islet beta cells in both rodents and humans. Co-localization of Cx36 with Cx30.2 has been recently identified. Functional studies in Cx36 deficient mice have provided direct evidence that Cx36 gap junction channels are necessary for the synchronization of [Ca(2+)]i oscillations in islet beta cells. The latter allows for the generation of insulin pulses in a single perfused islet. Moreover, Cx36 deficient mice were found to have altered serum insulin pulse dynamics and to be glucose intolerant. In addition, Cx36 has been recently identified as an early gene that is specifically expressed in embryonic beta cells, whose transcript and protein are upregulated in unison with the main wave of beta cell differentiation. In conclusion, Cx36 is critical for endocrine pancreatic function and may represent a molecular target for future prevention and treatment of diabetes. This article is part of the Special Issue Section entitled 'Current Pharmacology of Gap Junction Channels and Hemichannels'.
Collapse
Affiliation(s)
- E Martha Pérez-Armendariz
- Unidad de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Torre de Investigación 5to piso, Avenida Universidad 3000, Circuito Interior, Ciudad Universitaria, UNAM, México D.F. 04510, Mexico; Hospital General de México, Hospital General de México/Unidad de Medicina Experimental, Facultad de Medicina, UNAM, Dr Balmis 148, Colonia Doctores, Delegación Cuahutémoc, CP 06726 Ciudad de México, Mexico; Departamento of Biología Celular yTisular, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Circuito Interior, Ciudad Universitaria, UNAM, Mexico D.F. 04510, Mexico.
| |
Collapse
|
100
|
Meda P, Schuit F. Glucose-stimulated insulin secretion: the hierarchy of its multiple cellular and subcellular mechanisms. Diabetologia 2013; 56:2552-5. [PMID: 24114115 DOI: 10.1007/s00125-013-3073-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 09/12/2013] [Indexed: 02/08/2023]
Abstract
Glucose-stimulated insulin secretion is ensured by multiple molecular, cellular and tissue events. In this issue of Diabetologia, Low et al (DOI: 10.1007/s00125-013-3019-5 ) have taken an important new step towards understanding the hierarchical organisation of these events, by monitoring in vitro the individual exocytosis of multiple beta cells within intact mouse islets. The authors show that glucose stimulation markedly increases the number of exocytotic events per cell and, to a lesser extent, the number of beta cells contributing to this event. In this commentary we discuss these novel observations and propose that metabolic and electrical coupling of islet beta cells is responsible for a more homogeneous glucose-induced secretory response of cells in an intact islet as compared with isolated beta cells.
Collapse
Affiliation(s)
- Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva, School of Medicine, CMU, 1 rue Michel Servet, CH - 1211, Genève 4, Switzerland,
| | | |
Collapse
|