1
|
Moon DO. Interplay between paclitaxel, gap junctions, and kinases: unraveling mechanisms of action and resistance in cancer therapy. Mol Biol Rep 2024; 51:472. [PMID: 38551726 DOI: 10.1007/s11033-024-09411-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/01/2024] [Indexed: 04/02/2024]
Abstract
This comprehensive review elucidates the multifaceted roles of paclitaxel, a key chemotherapeutic agent, in cancer therapy, with a focus on its interactions with gap junctions and related kinases. Paclitaxel, with its complex diterpene structure, mediates its anticancer effects predominantly through specific interactions with β-tubulin, instigating cell cycle arrest and triggering various cell death pathways, including apoptosis, pyroptosis, ferroptosis, and necroptosis. The paper systematically delineates the chemical attributes and action mechanisms of paclitaxel and its analogs, underscoring their capacity to disrupt microtubule dynamics, thereby leading to mitotic arrest and subsequent cell death induction. It also scrutinizes the pivotal role of gap junctions, composed of connexin proteins, in the modulation of cancer cell behavior and chemoresistance, especially in the milieu of paclitaxel administration. The review articulates how gap junctions can either suppress tumors or contribute to cancer progression, thereby influencing chemotherapy outcomes. Furthermore, the paper provides an in-depth analysis of how paclitaxel modulates gap junction-associated kinases via phosphorylation, influencing the drug's therapeutic efficacy and resistance profiles. By integrating insights from numerous key studies, the review offers a comprehensive understanding of the interplay between paclitaxel, gap junctions, and kinases, shedding light on potential approaches to augment paclitaxel's anti-tumor effectiveness and counteract chemoresistance in cancer treatment.
Collapse
Affiliation(s)
- Dong-Oh Moon
- Department of Biology Education, Daegu University, 201, Daegudae-ro, Gyeongsan-si, Gyeongsangbuk-do, 38453, Republic of Korea.
| |
Collapse
|
2
|
Abstract
Gap junctions between neurons of the brain are thought to be present in only certain cell types, and they mostly connect dendrites, somata, and axons. Synapses with gap junctions serve bidirectional metabolic and electrical coupling between connected neuronal compartments. Although plasticity of electrical synapses has been described, recent evidence of the presence of silent, but activatable, gap junctions suggests that electrical nodes in a neuronal circuit can be added or suppressed by changes in the synaptic microenvironment. This opens the possibility of reconfiguration of neuronal ensembles in response to activity. Moreover, the coexistence of gap junctions in a glutamatergic synapse may add electric and metabolic coupling to a neuronal aggregate and may serve to constitute primed ensembles within a higher-order neural network. The interaction of chemical with electrical synapses should be further explored to find, especially, emerging properties of neuronal ensembles. It will be worth to reexamine in a new light the "functional" implications of the "anatomic" concepts: "continuity" and "contiguity," which were championed by Golgi and Ramón y Cajal, respectively. In any case, exploring the versatility of the gap junctions will likely enrich the heuristic aspects of the neural and network postulates.
Collapse
Affiliation(s)
- Rafael Gutiérrez
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| |
Collapse
|
3
|
St. Clair JR, Westacott MJ, Miranda J, Farnsworth NL, Kravets V, Schleicher WE, Dwulet JM, Levitt CH, Heintz A, Ludin NWF, Benninger RKP. Restoring connexin-36 function in diabetogenic environments precludes mouse and human islet dysfunction. J Physiol 2023; 601:4053-4072. [PMID: 37578890 PMCID: PMC10508056 DOI: 10.1113/jp282114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 07/27/2023] [Indexed: 08/16/2023] Open
Abstract
The secretion of insulin from β-cells in the islet of Langerhans is governed by a series of metabolic and electrical events, which can fail during the progression of type 2 diabetes (T2D). β-cells are electrically coupled via connexin-36 (Cx36) gap junction channels, which coordinates the pulsatile dynamics of [Ca2+ ] and insulin release across the islet. Factors such as pro-inflammatory cytokines and free fatty acids disrupt gap junction coupling under in vitro conditions. Here we test whether gap junction coupling and coordinated [Ca2+ ] dynamics are disrupted in T2D, and whether recovery of gap junction coupling can recover islet function. We examine islets from donors with T2D, from db/db mice, and islets treated with pro-inflammatory cytokines (TNF-α, IL-1β, IFN-ɣ) or free fatty acids (palmitate). We modulate gap junction coupling using Cx36 over-expression or pharmacological activation via modafinil. We also develop a peptide mimetic (S293) of the c-terminal regulatory site of Cx36 designed to compete against its phosphorylation. Cx36 gap junction permeability and [Ca2+ ] dynamics were disrupted in islets from both human donors with T2D and db/db mice, and in islets treated with pro-inflammatory cytokines or palmitate. Cx36 over-expression, modafinil treatment and S293 peptide all enhanced Cx36 gap junction coupling and protected against declines in coordinated [Ca2+ ] dynamics. Cx36 over-expression and S293 peptide also reduced apoptosis induced by pro-inflammatory cytokines. Critically, S293 peptide rescued gap junction coupling and [Ca2+ ] dynamics in islets from both db/db mice and a sub-set of T2D donors. Thus, recovering or enhancing Cx36 gap junction coupling can improve islet function in diabetes. KEY POINTS: Connexin-36 (Cx36) gap junction permeability and associated coordination of [Ca2+ ] dynamics is diminished in human type 2 diabetes (T2D) and mouse models of T2D. Enhancing Cx36 gap junction permeability protects against disruptions to the coordination of [Ca2+ ] dynamics. A novel peptide mimetic of the Cx36 c-terminal regulatory region protects against declines in Cx36 gap junction permeability. Pharmacological elevation in Cx36 or Cx36 peptide mimetic recovers [Ca2+ ] dynamics and glucose-stimulated insulin secretion in human T2D and mouse models of T2D.
Collapse
Affiliation(s)
- Joshua R St. Clair
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Matthew J Westacott
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Jose Miranda
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Nikki L Farnsworth
- Barbara Davis Center for Diabetes, University of Colorado
Denver | Anschutz Medical Campus, Aurora, CO
| | - Vira Kravets
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Wolfgang E Schleicher
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - JaeAnn M Dwulet
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Claire H Levitt
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Audrey Heintz
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Nurin WF Ludin
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Richard KP Benninger
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
- Barbara Davis Center for Diabetes, University of Colorado
Denver | Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
4
|
Skelin Klemen M, Dolenšek J, Križančić Bombek L, Pohorec V, Gosak M, Slak Rupnik M, Stožer A. The effect of forskolin and the role of Epac2A during activation, activity, and deactivation of beta cell networks. Front Endocrinol (Lausanne) 2023; 14:1225486. [PMID: 37701894 PMCID: PMC10494243 DOI: 10.3389/fendo.2023.1225486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/08/2023] [Indexed: 09/14/2023] Open
Abstract
Beta cells couple stimulation by glucose with insulin secretion and impairments in this coupling play a central role in diabetes mellitus. Cyclic adenosine monophosphate (cAMP) amplifies stimulus-secretion coupling via protein kinase A and guanine nucleotide exchange protein 2 (Epac2A). With the present research, we aimed to clarify the influence of cAMP-elevating diterpene forskolin on cytoplasmic calcium dynamics and intercellular network activity, which are two of the crucial elements of normal beta cell stimulus-secretion coupling, and the role of Epac2A under normal and stimulated conditions. To this end, we performed functional multicellular calcium imaging of beta cells in mouse pancreas tissue slices after stimulation with glucose and forskolin in wild-type and Epac2A knock-out mice. Forskolin evoked calcium signals in otherwise substimulatory glucose and beta cells from Epac2A knock-out mice displayed a faster activation. During the plateau phase, beta cells from Epac2A knock-out mice displayed a slightly higher active time in response to glucose compared with wild-type littermates, and stimulation with forskolin increased the active time via an increase in oscillation frequency and a decrease in oscillation duration in both Epac2A knock-out and wild-type mice. Functional network properties during stimulation with glucose did not differ in Epac2A knock-out mice, but the presence of Epac2A was crucial for the protective effect of stimulation with forskolin in preventing a decline in beta cell functional connectivity with time. Finally, stimulation with forskolin prolonged beta cell activity during deactivation, especially in Epac2A knock-out mice.
Collapse
Affiliation(s)
- Maša Skelin Klemen
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Jurij Dolenšek
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | | | - Viljem Pohorec
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Marko Gosak
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
- Alma Mater Europaea, European Center Maribor, Maribor, Slovenia
| | - Marjan Slak Rupnik
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Alma Mater Europaea, European Center Maribor, Maribor, Slovenia
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Andraž Stožer
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| |
Collapse
|
5
|
Kovács-Öller T, Szarka G, Hoffmann G, Péntek L, Valentin G, Ross L, Völgyi B. Extrinsic and Intrinsic Factors Determine Expression Levels of Gap Junction-Forming Connexins in the Mammalian Retina. Biomolecules 2023; 13:1119. [PMID: 37509155 PMCID: PMC10377540 DOI: 10.3390/biom13071119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Gap junctions (GJs) are not static bridges; instead, GJs as well as the molecular building block connexin (Cx) proteins undergo major expression changes in the degenerating retinal tissue. Various progressive diseases, including retinitis pigmentosa, glaucoma, age-related retinal degeneration, etc., affect neurons of the retina and thus their neuronal connections endure irreversible changes as well. Although Cx expression changes might be the hallmarks of tissue deterioration, GJs are not static bridges and as such they undergo adaptive changes even in healthy tissue to respond to the ever-changing environment. It is, therefore, imperative to determine these latter adaptive changes in GJ functionality as well as in their morphology and Cx makeup to identify and distinguish them from alterations following tissue deterioration. In this review, we summarize GJ alterations that take place in healthy retinal tissue and occur on three different time scales: throughout the entire lifespan, during daily changes and as a result of quick changes of light adaptation.
Collapse
Affiliation(s)
- Tamás Kovács-Öller
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
- Department of Neurobiology, University of Pécs, 7624 Pécs, Hungary
- NEURON-066 Rethealthsi Research Group, 7624 Pécs, Hungary
- Center for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| | - Gergely Szarka
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
- Department of Neurobiology, University of Pécs, 7624 Pécs, Hungary
- NEURON-066 Rethealthsi Research Group, 7624 Pécs, Hungary
- Center for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| | - Gyula Hoffmann
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
- Department of Neurobiology, University of Pécs, 7624 Pécs, Hungary
- NEURON-066 Rethealthsi Research Group, 7624 Pécs, Hungary
- Center for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| | - Loretta Péntek
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
- Department of Neurobiology, University of Pécs, 7624 Pécs, Hungary
| | - Gréta Valentin
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
- Department of Neurobiology, University of Pécs, 7624 Pécs, Hungary
| | - Liliana Ross
- Faculty of Science, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Béla Völgyi
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
- Department of Neurobiology, University of Pécs, 7624 Pécs, Hungary
- NEURON-066 Rethealthsi Research Group, 7624 Pécs, Hungary
- Center for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| |
Collapse
|
6
|
Maruyama T, Ishii T, Kaneda M. Starburst amacrine cells form gap junctions in the early postnatal stage of the mouse retina. Front Cell Neurosci 2023; 17:1173579. [PMID: 37293630 PMCID: PMC10244514 DOI: 10.3389/fncel.2023.1173579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/03/2023] [Indexed: 06/10/2023] Open
Abstract
Although gap junctional coupling in the developing retina is important for the maturation of neuronal networks, its role in the development of individual neurons remains unclear. Therefore, we herein investigated whether gap junctional coupling by starburst amacrine cells (SACs), a key neuron for the formation of direction selectivity, occurs during the developmental stage in the mouse retina. Neurobiotin-injected SACs coupled with many neighboring cells before eye-opening. The majority of tracer-coupled cells were retinal ganglion cells, and tracer coupling was not detected between SACs. The number of tracer-coupled cells significantly decreased after eye-opening and mostly disappeared by postnatal day 28 (P28). Membrane capacitance (Cm), an indicator of the formation of electrical coupling with gap junctions, was larger in SACs before than after eye-opening. The application of meclofenamic acid, a gap junction blocker, reduced the Cm of SACs. Gap junctional coupling by SACs was regulated by dopamine D1 receptors before eye-opening. In contrast, the reduction in gap junctional coupling after eye-opening was not affected by visual experience. At the mRNA level, 4 subtypes of connexins (23, 36, 43, and 45) were detected in SACs before eye-opening. Connexin 43 expression levels significantly decreased after eye-opening. These results indicate that gap junctional coupling by SACs occurs during the developmental period and suggest that the elimination of gap junctions proceeds with the innate system.
Collapse
|
7
|
Myles WE, McFadden SA. Analytical methods for assessing retinal cell coupling using cut-loading. PLoS One 2022; 17:e0271744. [PMID: 35853039 PMCID: PMC9295955 DOI: 10.1371/journal.pone.0271744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 07/06/2022] [Indexed: 11/25/2022] Open
Abstract
Electrical coupling between retinal neurons contributes to the functional complexity of visual circuits. “Cut-loading” methods allow simultaneous assessment of cell-coupling between multiple retinal cell-types, but existing analysis methods impede direct comparison with gold standard direct dye injection techniques. In the current study, we both improved an existing method and developed two new approaches to address observed limitations. Each method of analysis was applied to cut-loaded dark-adapted Guinea pig retinae (n = 29) to assess coupling strength in the axonless horizontal cell type (‘a-type’, aHCs). Method 1 was an improved version of the standard protocol and described the distance of dye-diffusion (space constant). Method 2 adjusted for the geometric path of dye-transfer through cut-loaded cells and extracted the rate of dye-transfer across gap-junctions in terms of the coupling coefficient (kj). Method 3 measured the diffusion coefficient (De) perpendicular to the cut-axis. Dye transfer was measured after one of five diffusion times (1–20 mins), or with a coupling inhibitor, meclofenamic acid (MFA) (50–500μM after 20 mins diffusion). The standard protocol fits an exponential decay function to the fluorescence profile of a specified retina layer but includes non-specific background fluorescence. This was improved by measuring the fluorescence of individual cell soma and excluding from the fit non-horizontal cells located at the cut-edge (p<0.001) (Method 1). The space constant (Method 1) increased with diffusion time (p<0.01), whereas Methods 2 (p = 0.54) and 3 (p = 0.63) produced consistent results across all diffusion times. Adjusting distance by the mean cell-cell spacing within each tissue reduced the incidence of outliers across all three methods. Method 1 was less sensitive to detecting changes induced by MFA than Methods 2 (p<0.01) and 3 (p<0.01). Although the standard protocol was easily improved (Method 1), Methods 2 and 3 proved more sensitive and generalisable; allowing for detailed assessment of the tracer kinetics between different populations of gap-junction linked cell networks and direct comparison to dye-injection techniques.
Collapse
Affiliation(s)
- William E. Myles
- College of Engineering, Science and Environment, University of Newcastle, Callaghan, NSW, Australia
- * E-mail:
| | - Sally A. McFadden
- College of Engineering, Science and Environment, University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
8
|
Glycine Release Is Potentiated by cAMP via EPAC2 and Ca 2+ Stores in a Retinal Interneuron. J Neurosci 2021; 41:9503-9520. [PMID: 34620721 PMCID: PMC8612479 DOI: 10.1523/jneurosci.0670-21.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 11/21/2022] Open
Abstract
Neuromodulation via the intracellular second messenger cAMP is ubiquitous at presynaptic nerve terminals. This modulation of synaptic transmission allows exocytosis to adapt to stimulus levels and reliably encode information. The AII amacrine cell (AII-AC) is a central hub for signal processing in the mammalian retina. The main apical dendrite of the AII-AC is connected to several lobular appendages that release glycine onto OFF cone bipolar cells and ganglion cells. However, the influence of cAMP on glycine release is not well understood. Using membrane capacitance measurements from mouse AII-ACs to directly measure exocytosis, we observe that intracellular dialysis of 1 mm cAMP enhances exocytosis without affecting the L-type Ca2+ current. Responses to depolarizing pulses of various durations show that the size of the readily releasable pool of vesicles nearly doubles with cAMP, while paired-pulse depression experiments suggest that release probability does not change. Specific agonists and antagonists for exchange protein activated by cAMP 2 (EPAC2) revealed that the cAMP-induced enhancement of exocytosis requires EPAC2 activation. Furthermore, intact Ca2+ stores were also necessary for the cAMP potentiation of exocytosis. Postsynaptic recordings from OFF cone bipolar cells showed that increasing cAMP with forskolin potentiated the frequency of glycinergic spontaneous IPSCs. We propose that cAMP elevations in the AII-AC lead to a robust enhancement of glycine release through an EPAC2 and Ca2+ store signaling pathway. Our results thus contribute to a better understanding of how AII-AC crossover inhibitory circuits adapt to changes in ambient luminance.SIGNIFICANCE STATEMENT The mammalian retina operates over a wide dynamic range of light intensities and contrast levels. To optimize the signal-to-noise ratio of processed visual information, both excitatory and inhibitory synapses within the retina must modulate their gain in synaptic transmission to adapt to different levels of ambient light. Here we show that increases of cAMP concentration within AII amacrine cells produce enhanced exocytosis from these glycinergic interneurons. Therefore, we propose that light-sensitive neuromodulators may change the output of glycine release from AII amacrine cells. This novel mechanism may fine-tune the amount of tonic and phasic synaptic inhibition received by bipolar cell terminals and, consequently, the spiking patterns that ganglion cells send to the upstream visual areas of the brain.
Collapse
|
9
|
Stožer A, Paradiž Leitgeb E, Pohorec V, Dolenšek J, Križančić Bombek L, Gosak M, Skelin Klemen M. The Role of cAMP in Beta Cell Stimulus-Secretion and Intercellular Coupling. Cells 2021; 10:1658. [PMID: 34359828 PMCID: PMC8304079 DOI: 10.3390/cells10071658] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/18/2021] [Accepted: 06/28/2021] [Indexed: 12/22/2022] Open
Abstract
Pancreatic beta cells secrete insulin in response to stimulation with glucose and other nutrients, and impaired insulin secretion plays a central role in development of diabetes mellitus. Pharmacological management of diabetes includes various antidiabetic drugs, including incretins. The incretin hormones, glucagon-like peptide-1 and gastric inhibitory polypeptide, potentiate glucose-stimulated insulin secretion by binding to G protein-coupled receptors, resulting in stimulation of adenylate cyclase and production of the secondary messenger cAMP, which exerts its intracellular effects through activation of protein kinase A or the guanine nucleotide exchange protein 2A. The molecular mechanisms behind these two downstream signaling arms are still not fully elucidated and involve many steps in the stimulus-secretion coupling cascade, ranging from the proximal regulation of ion channel activity to the central Ca2+ signal and the most distal exocytosis. In addition to modifying intracellular coupling, the effect of cAMP on insulin secretion could also be at least partly explained by the impact on intercellular coupling. In this review, we systematically describe the possible roles of cAMP at these intra- and inter-cellular signaling nodes, keeping in mind the relevance for the whole organism and translation to humans.
Collapse
Affiliation(s)
- Andraž Stožer
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; (A.S.); (E.P.L.); (V.P.); (J.D.); (L.K.B.); (M.G.)
| | - Eva Paradiž Leitgeb
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; (A.S.); (E.P.L.); (V.P.); (J.D.); (L.K.B.); (M.G.)
| | - Viljem Pohorec
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; (A.S.); (E.P.L.); (V.P.); (J.D.); (L.K.B.); (M.G.)
| | - Jurij Dolenšek
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; (A.S.); (E.P.L.); (V.P.); (J.D.); (L.K.B.); (M.G.)
- Faculty of Natural Sciences and Mathematics, University of Maribor, SI-2000 Maribor, Slovenia
| | - Lidija Križančić Bombek
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; (A.S.); (E.P.L.); (V.P.); (J.D.); (L.K.B.); (M.G.)
| | - Marko Gosak
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; (A.S.); (E.P.L.); (V.P.); (J.D.); (L.K.B.); (M.G.)
- Faculty of Natural Sciences and Mathematics, University of Maribor, SI-2000 Maribor, Slovenia
| | - Maša Skelin Klemen
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; (A.S.); (E.P.L.); (V.P.); (J.D.); (L.K.B.); (M.G.)
| |
Collapse
|
10
|
Chen AM, Azar SS, Harris A, Brecha NC, Pérez de Sevilla Müller L. PTEN Expression Regulates Gap Junction Connectivity in the Retina. Front Neuroanat 2021; 15:629244. [PMID: 34093139 PMCID: PMC8172595 DOI: 10.3389/fnana.2021.629244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/26/2021] [Indexed: 12/16/2022] Open
Abstract
Manipulation of the phosphatase and tensin homolog (PTEN) pathway has been suggested as a therapeutic approach to treat or prevent vision loss due to retinal disease. In this study, we investigated the effects of deleting one copy of Pten in a well-characterized class of retinal ganglion cells called α-ganglion cells in the mouse retina. In Pten +/- retinas, α-ganglion cells did not exhibit major changes in their dendritic structure, although most cells developed a few, unusual loop-forming dendrites. By contrast, α-ganglion cells exhibited a significant decrease in heterologous and homologous gap junction mediated cell coupling with other retinal ganglion and amacrine cells. Additionally, the majority of OFF α-ganglion cells (12/18 cells) formed novel coupling to displaced amacrine cells. The number of connexin36 puncta, the predominant connexin that mediates gap junction communication at electrical synapses, was decreased by at least 50% on OFF α-ganglion cells. Reduced and incorrect gap junction connectivity of α-ganglion cells will affect their functional properties and alter visual image processing in the retina. The anomalous connectivity of retinal ganglion cells would potentially limit future therapeutic approaches involving manipulation of the Pten pathway for treating ganglion cell degeneration in diseases like glaucoma, traumatic brain injury, Parkinson's, and Alzheimer's diseases.
Collapse
Affiliation(s)
- Ashley M. Chen
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California, Los Angeles, Los Angeles, CA, United States
| | - Shaghauyegh S. Azar
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California, Los Angeles, Los Angeles, CA, United States
| | - Alexander Harris
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California, Los Angeles, Los Angeles, CA, United States
| | - Nicholas C. Brecha
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California, Los Angeles, Los Angeles, CA, United States
- Stein Eye Institute, David Geffen School of Medicine at Los Angeles, University of California, Los Angeles, Los Angeles, CA, United States
- CURE Digestive Diseases Research Center, David Geffen School of Medicine at Los Angeles, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Administration Greater Los Angeles Health System, Los Angeles, CA, United States
| | - Luis Pérez de Sevilla Müller
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
11
|
Zoidl GR, Spray DC. The Roles of Calmodulin and CaMKII in Cx36 Plasticity. Int J Mol Sci 2021; 22:4473. [PMID: 33922931 PMCID: PMC8123330 DOI: 10.3390/ijms22094473] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/17/2021] [Accepted: 04/20/2021] [Indexed: 01/07/2023] Open
Abstract
Anatomical and electrophysiological evidence that gap junctions and electrical coupling occur between neurons was initially confined to invertebrates and nonmammals and was thought to be a primitive form of synaptic transmission. More recent studies revealed that electrical communication is common in the mammalian central nervous system (CNS), often coexisting with chemical synaptic transmission. The subsequent progress indicated that electrical synapses formed by the gap junction protein connexin-36 (Cx36) and its paralogs in nonmammals constitute vital elements in mammalian and fish synaptic circuitry. They govern the collective activity of ensembles of coupled neurons, and Cx36 gap junctions endow them with enormous adaptive plasticity, like that seen at chemical synapses. Moreover, they orchestrate the synchronized neuronal network activity and rhythmic oscillations that underlie the fundamental integrative processes, such as memory and learning. Here, we review the available mechanistic evidence and models that argue for the essential roles of calcium, calmodulin, and the Ca2+/calmodulin-dependent protein kinase II in integrating calcium signals to modulate the strength of electrical synapses through interactions with the gap junction protein Cx36.
Collapse
Affiliation(s)
- Georg R. Zoidl
- Department of Biology & Center for Vision Research (CVR), York University, Toronto, ON M3J 1P3, Canada
| | - David C. Spray
- Dominick P. Purpura Department of Neuroscience & Department of Medicine (Cardiology), Albert Einstein College of Medicine, New York, NY 10461, USA;
| |
Collapse
|
12
|
Exacerbation of Epilepsy by Astrocyte Alkalization and Gap Junction Uncoupling. J Neurosci 2021; 41:2106-2118. [PMID: 33478985 DOI: 10.1523/jneurosci.2365-20.2020] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/02/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022] Open
Abstract
Seizures invite seizures. At the initial stage of epilepsy, seizures intensify with each episode; however, the mechanisms underlying this exacerbation remain to be solved. Astrocytes have a strong control over neuronal excitability and the mode of information processing. This control is accomplished by adjusting the levels of various ions in the extracellular space. The network of astrocytes connected via gap junctions allows a wider or more confined distribution of these ions depending on the open probability of the gap junctions. K+ clearance relies on the K+ uptake by astrocytes and the subsequent diffusion of K+ through the astrocyte network. When astrocytes become uncoupled, K+ clearance becomes hindered. Accumulation of extracellular K+ leads to hyperexcitability of neurons. Here, using acute hippocampal slices from mice, we uncovered that brief periods of epileptiform activity result in gap junction uncoupling. In slices that experienced short-term epileptiform activity, extracellular K+ transients in response to glutamate became prolonged. Na+ imaging with a fluorescent indicator indicated that intercellular diffusion of small cations in the astrocytic syncytium via gap junctions became rapidly restricted after epileptiform activity. Using a transgenic mouse with astrocyte-specific expression of a pH sensor (Lck-E2GFP), we confirmed that astrocytes react to epileptiform activity with intracellular alkalization. Application of Na+/HCO3 - cotransporter blocker led to the suppression of intracellular alkalization of astrocytes and to the prevention of astrocyte uncoupling and hyperactivity intensification both in vitro and in vivo Therefore, the inhibition of astrocyte alkalization could become a promising therapeutic strategy for countering epilepsy development.SIGNIFICANCE STATEMENT We aimed to understand the mechanisms underlying the plastic change of forebrain circuits associated with the intensification of epilepsy. Here, we demonstrate that first-time exposure to only brief periods of epileptiform activity results in acute disturbance of the intercellular astrocyte network formed by gap junctions in hippocampal tissue slices from mice. Moreover, rapid clearance of K+ from the extracellular space was impaired. Epileptiform activity activated inward Na+/HCO3 - cotransport in astrocytes by cell depolarization, resulting in their alkalization. Our data suggest that alkaline pH shifts in astrocytes lead to gap junction uncoupling, hampering K+ clearance, and thereby to exacerbation of epilepsy. Pharmacological intervention could become a promising new strategy to dampen neuronal hyperexcitability and epileptogenesis.
Collapse
|
13
|
Landis EG, Park HN, Chrenek M, He L, Sidhu C, Chakraborty R, Strickland R, Iuvone PM, Pardue MT. Ambient Light Regulates Retinal Dopamine Signaling and Myopia Susceptibility. Invest Ophthalmol Vis Sci 2021; 62:28. [PMID: 33502461 PMCID: PMC7846952 DOI: 10.1167/iovs.62.1.28] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purpose Exposure to high-intensity or outdoor lighting has been shown to decrease the severity of myopia in both human epidemiological studies and animal models. Currently, it is not fully understood how light interacts with visual signaling to impact myopia. Previous work performed in the mouse retina has demonstrated that functional rod photoreceptors are needed to develop experimentally-induced myopia, alluding to an essential role for rod signaling in refractive development. Methods To determine whether dim rod-dominated illuminance levels influence myopia susceptibility, we housed male C57BL/6J mice under 12:12 light/dark cycles with scotopic (1.6 × 10−3 candela/m2), mesopic (1.6 × 101 cd/m2), or photopic (4.7 × 103 cd/m2) lighting from post-natal day 23 (P23) to P38. Half the mice received monocular exposure to −10 diopter (D) lens defocus from P28–38. Molecular assays to measure expression and content of DA-related genes and protein were conducted to determine how illuminance and lens defocus alter dopamine (DA) synthesis, storage, uptake, and degradation and affect myopia susceptibility in mice. Results We found that mice exposed to either scotopic or photopic lighting developed significantly less severe myopic refractive shifts (lens treated eye minus contralateral eye; –1.62 ± 0.37D and −1.74 ± 0.44D, respectively) than mice exposed to mesopic lighting (–3.61 ± 0.50D; P < 0.005). The 3,4-dihydroxyphenylacetic acid /DA ratio, indicating DA activity, was highest under photopic light regardless of lens defocus treatment (controls: 0.09 ± 0.011 pg/mg, lens defocus: 0.08 ± 0.008 pg/mg). Conclusions Lens defocus interacted with ambient conditions to differentially alter myopia susceptibility and DA-related genes and proteins. Collectively, these results show that scotopic and photopic lighting protect against lens-induced myopia, potentially indicating that a broad range of light levels are important in refractive development.
Collapse
Affiliation(s)
- Erica G Landis
- Department of Neuroscience, Emory University, Atlanta, Georgia, United States.,Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Atlanta, Georgia, United States
| | - Han Na Park
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Micah Chrenek
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Li He
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Curran Sidhu
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Ranjay Chakraborty
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States.,Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Atlanta, Georgia, United States
| | - Ryan Strickland
- Department of Neuroscience, Emory University, Atlanta, Georgia, United States
| | - P Michael Iuvone
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States.,Department of Pharmacology, Emory University, Atlanta, Georgia, United States
| | - Machelle T Pardue
- Department of Neuroscience, Emory University, Atlanta, Georgia, United States.,Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Atlanta, Georgia, United States.,Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, United States
| |
Collapse
|
14
|
Tetenborg S, Wang HY, Nemitz L, Depping A, Espejo AB, Aseervatham J, Bedford MT, Janssen-Bienhold U, O'Brien J, Dedek K. Phosphorylation of Connexin36 near the C-terminus switches binding affinities for PDZ-domain and 14-3-3 proteins in vitro. Sci Rep 2020; 10:18378. [PMID: 33110101 PMCID: PMC7592057 DOI: 10.1038/s41598-020-75375-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 10/12/2020] [Indexed: 12/20/2022] Open
Abstract
Connexin36 (Cx36) is the most abundant connexin in central nervous system neurons. It forms gap junction channels that act as electrical synapses. Similar to chemical synapses, Cx36-containing gap junctions undergo activity-dependent plasticity and complex regulation. Cx36 gap junctions represent multimolecular complexes and contain cytoskeletal, regulatory and scaffolding proteins, which regulate channel conductance, assembly and turnover. The amino acid sequence of mammalian Cx36 harbors a phosphorylation site for the Ca2+/calmodulin-dependent kinase II at serine 315. This regulatory site is homologous to the serine 298 in perch Cx35 and in close vicinity to a PDZ binding domain at the very C-terminal end of the protein. We hypothesized that this phosphorylation site may serve as a molecular switch, influencing the affinity of the PDZ binding domain for its binding partners. Protein microarray and pulldown experiments revealed that this is indeed the case: phosphorylation of serine 298 decreased the binding affinity for MUPP1, a known scaffolding partner of connexin36, and increased the binding affinity for two different 14-3-3 proteins. Although we did not find the same effect in cell culture experiments, our data suggest that phosphorylation of serine 315/298 may serve to recruit different proteins to connexin36/35-containing gap junctions in an activity-dependent manner.
Collapse
Affiliation(s)
- Stephan Tetenborg
- Animal Navigation/Neurosensorics, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany
- Ruiz Department of Ophthalmology & Visual Science, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Helen Y Wang
- Ruiz Department of Ophthalmology & Visual Science, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Lena Nemitz
- Visual Neuroscience, Dept. of Neuroscience, University of Oldenburg, Oldenburg, Germany
| | - Anne Depping
- Animal Navigation/Neurosensorics, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany
| | - Alexsandra B Espejo
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Smithville, TX, 78957, USA
| | - Jaya Aseervatham
- Ruiz Department of Ophthalmology & Visual Science, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Mark T Bedford
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Smithville, TX, 78957, USA
| | - Ulrike Janssen-Bienhold
- Visual Neuroscience, Dept. of Neuroscience, University of Oldenburg, Oldenburg, Germany
- Research Center Neurosensory Science, University of Oldenburg, Oldenburg, Germany
| | - John O'Brien
- Ruiz Department of Ophthalmology & Visual Science, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Karin Dedek
- Animal Navigation/Neurosensorics, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany.
- Research Center Neurosensory Science, University of Oldenburg, Oldenburg, Germany.
| |
Collapse
|
15
|
Aseervatham J, Li X, Mitchell CK, Lin YP, Heidelberger R, O’Brien J. Calmodulin Binding to Connexin 35: Specializations to Function as an Electrical Synapse. Int J Mol Sci 2020; 21:E6346. [PMID: 32882943 PMCID: PMC7504508 DOI: 10.3390/ijms21176346] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 08/27/2020] [Accepted: 08/30/2020] [Indexed: 12/25/2022] Open
Abstract
Calmodulin binding is a nearly universal property of gap junction proteins, imparting a calcium-dependent uncoupling behavior that can serve in an emergency to decouple a stressed cell from its neighbors. However, gap junctions that function as electrical synapses within networks of neurons routinely encounter large fluctuations in local cytoplasmic calcium concentration; frequent uncoupling would be impractical and counterproductive. We have studied the properties and functional consequences of calmodulin binding to the electrical synapse protein Connexin 35 (Cx35 or gjd2b), homologous to mammalian Connexin 36 (Cx36 or gjd2). We find that specializations in Cx35 calmodulin binding sites make it relatively impervious to moderately high levels of cytoplasmic calcium. Calmodulin binding to a site in the C-terminus causes uncoupling when calcium reaches low micromolar concentrations, a behavior prevented by mutations that eliminate calmodulin binding. However, milder stimuli promote calcium/calmodulin-dependent protein kinase II activity that potentiates coupling without interference from calmodulin binding. A second calmodulin binding site in the end of the Cx35 cytoplasmic loop, homologous to a calmodulin binding site present in many connexins, binds calmodulin with very low affinity and stoichiometry. Together, the calmodulin binding sites cause Cx35 to uncouple only at extreme levels of intracellular calcium.
Collapse
Affiliation(s)
- Jaya Aseervatham
- Ruiz Department of Ophthalmology & Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.A.); (X.L.); (C.K.M.); (Y.-P.L.)
| | - Xiaofan Li
- Ruiz Department of Ophthalmology & Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.A.); (X.L.); (C.K.M.); (Y.-P.L.)
| | - Cheryl K. Mitchell
- Ruiz Department of Ophthalmology & Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.A.); (X.L.); (C.K.M.); (Y.-P.L.)
| | - Ya-Ping Lin
- Ruiz Department of Ophthalmology & Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.A.); (X.L.); (C.K.M.); (Y.-P.L.)
| | - Ruth Heidelberger
- Department of Neurobiology & Anatomy, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
- The MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - John O’Brien
- Ruiz Department of Ophthalmology & Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.A.); (X.L.); (C.K.M.); (Y.-P.L.)
- The MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
16
|
Banerjee S, Wang Q, Zhao F, Tang G, So C, Tse D, To CH, Feng Y, Zhou X, Pan F. Increased Connexin36 Phosphorylation in AII Amacrine Cell Coupling of the Mouse Myopic Retina. Front Cell Neurosci 2020; 14:124. [PMID: 32547367 PMCID: PMC7278884 DOI: 10.3389/fncel.2020.00124] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 04/15/2020] [Indexed: 12/14/2022] Open
Abstract
Myopia is a substantial public health problem worldwide. In the myopic retina, distant images are focused in front of the photoreceptors. The cells and mechanisms for retinal signaling that account either for emmetropization (i.e., normal refraction) or for refractive errors have remained elusive. Gap junctions play a key component in enhancement of signal transmission in visual pathways. AII amacrine cells (ACs), coupled by connexin36, segregate signals into ON and OFF pathways. Coupling between AII ACs is actively modulated through phosphorylation at serine 293 via dopamine in the mouse retina. In this study, form deprivation mouse myopia models were used to evaluate the expression patterns of connexin36-positive plaques (structural assay) and the state of connexin36 phosphorylation (functional assay) in AII ACs, which was green fluorescent protein-expressing in the Fam81a mouse line. Single-cell RNA sequencing showed dopaminergic synapse and gap junction pathways of AII ACs were downregulated in the myopic retina, although Gjd2 mRNA expression remained the same. Compared with the normal refractive eye, phosphorylation of connexin36 was increased in the myopic retina, but expression of connexin36 remained unchanged. This increased phosphorylation of Cx36 could indicate increased functional gap junction coupling of AII ACs in the myopic retina, a possible adaptation to adjust to the altered noisy signaling status.
Collapse
Affiliation(s)
- Seema Banerjee
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Qin Wang
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Fuxin Zhao
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, The State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, China
| | - George Tang
- School of Clinical Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Chunghim So
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Dennis Tse
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Chi-Ho To
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Yun Feng
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
| | - Xiangtian Zhou
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, The State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, China
| | - Feng Pan
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| |
Collapse
|
17
|
Smedowski A, Akhtar S, Liu X, Pietrucha‐Dutczak M, Podracka L, Toropainen E, Alkanaan A, Ruponen M, Urtti A, Varjosalo M, Kaarniranta K, Lewin‐Kowalik J. Electrical synapses interconnecting axons revealed in the optic nerve head - a novel model of gap junctions' involvement in optic nerve function. Acta Ophthalmol 2020; 98:408-417. [PMID: 31602808 PMCID: PMC7318195 DOI: 10.1111/aos.14272] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 09/14/2019] [Indexed: 01/14/2023]
Abstract
PURPOSE To characterize newly discovered electrical synapses, formed by connexin (Cx) 36 and 45, between neighbouring axons within the optic nerve head. METHODS Twenty-five Wistar rats were killed by CO2 inhalation. Proximal and distal optic nerve (ON) stumps were collected and processed for immunostainings, electron microscopy (EM) with immunogold labelling, PCR and Western blots (WB). Additional 15 animals were deeply anaesthetized, and flash visual evoked potentials (fVEP) after retrobulbar injection of saline (negative control) or 100 μm meclofenamic acid solution (gap junctions' blocker) were recorded. Human paraffin cross-sections of eyeballs for immunostainings were obtained from the Human Eye Biobank for Research. RESULTS Immunostainings of both rat and human ON revealed the presence of Cx45 and 36 colocalizing with β3-tubulin, but not with glial fibrillary acidic protein (GFAP). In WB, Cx36 content in optic nerve was approximately halved when compared with retina (0.58 ± 0.005 in proximal stump and 0.44 ± 0.02 in distal stump), Cx45 showed higher levels (0.68 ± 0.01 in proximal stump and 0.9 ± 0.07 in distal stump). In immunogold-EM of optic nerve sections, we found electric synapses (formed mostly by Cx45) directly coupling neighbouring axons. In fVEP, blocking of gap junctions with meclofenamic acid resulted in significant prolongation of the latency of P1 wave up to 160% after 30 min (p < 0.001). CONCLUSIONS Optic nerve (ON) axons are equipped with electrical synapses composed of neuronal connexins, especially Cx45, creating direct morphological and functional connections between each other. This finding could have substantial implications for understanding of the pathogenesis of various optic neuropathies and identifies a new potential target for a therapeutic approach.
Collapse
Affiliation(s)
- Adrian Smedowski
- Department of PhysiologySchool of Medicine in KatowiceMedical University of SilesiaKatowicePoland
| | - Saeed Akhtar
- Department of OptometryCollege of Applied Medical SciencesKing Saud UniversityRiyadhKingdom of Saudi Arabia
| | - Xiaonan Liu
- Institute of BiotechnologyUniversity of HelsinkiHelsinkiFinland
| | - Marita Pietrucha‐Dutczak
- Department of PhysiologySchool of Medicine in KatowiceMedical University of SilesiaKatowicePoland
| | - Lucia Podracka
- School of PharmacyUniversity of Eastern FinlandKuopioFinland
| | | | - Aljoharah Alkanaan
- Department of OptometryCollege of Applied Medical SciencesKing Saud UniversityRiyadhKingdom of Saudi Arabia
| | - Marika Ruponen
- School of PharmacyUniversity of Eastern FinlandKuopioFinland
| | - Arto Urtti
- School of PharmacyUniversity of Eastern FinlandKuopioFinland
| | | | - Kai Kaarniranta
- Department of OphthalmologyUniversity of Eastern FinlandKuopioFinland,Department of OphthalmologyKuopio University HospitalKuopioFinland
| | - Joanna Lewin‐Kowalik
- Department of PhysiologySchool of Medicine in KatowiceMedical University of SilesiaKatowicePoland
| |
Collapse
|
18
|
Xie J, Wang X, Ge H, Peng F, Zheng N, Wang Q, Tao L. Cx32 mediates norepinephrine-promoted EGFR-TKI resistance in a gap junction-independent manner in non-small-cell lung cancer. J Cell Physiol 2019; 234:23146-23159. [PMID: 31152452 DOI: 10.1002/jcp.28881] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 05/07/2019] [Accepted: 05/09/2019] [Indexed: 01/10/2023]
Abstract
The second-generation EGFR-TKI Afatinib is an irreversible ErbB family blocker used to treat patients with non-small-cell lung cancer (NSCLC). Unfortunately, resistance to this drug develops over time, and patients are always under great psychological pressure. A previous study showed that chronic stress hormones participate in EGFR-TKI resistance via β2 -AR signaling via an IL-6 dependent mechanism. Our study further explores a novel potential underlying mechanism. In the present study, we show that the stress hormone norepinephrine (NE) promotes Afatinib resistance by upregulating Cx32 expression. Furthermore, we, for the first time, find that Cx32 is a target gene for transcription factor CREB and NE enhances Cx32 mRNA expression by activation of CREB. We also demonstrate that Cx32 promotes Afatinib resistance by decreasing the degradation of EGFR-TKI resistance-associated proteins (MET, IGF-1R) and by increasing their transcription levels. Together, these results reveal that the stress hormone NE accelerates Afatinib resistance by increasing the expression of Cx32, which augments MET and IGF-1R levels in cancer cells and provides a promising therapeutic strategy against EGFR-TKI Afatinib resistance in NSCLC.
Collapse
Affiliation(s)
- Jie Xie
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Xiyan Wang
- Tumor Research Institute, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, Xinjiang, China
| | - Hui Ge
- Tumor Research Institute, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, Xinjiang, China
| | - Fuhua Peng
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Ningze Zheng
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Qin Wang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Liang Tao
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
19
|
Brown CA, Del Corsso C, Zoidl C, Donaldson LW, Spray DC, Zoidl G. Tubulin-Dependent Transport of Connexin-36 Potentiates the Size and Strength of Electrical Synapses. Cells 2019; 8:E1146. [PMID: 31557934 PMCID: PMC6829524 DOI: 10.3390/cells8101146] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/17/2019] [Accepted: 09/20/2019] [Indexed: 12/12/2022] Open
Abstract
Connexin-36 (Cx36) electrical synapses strengthen transmission in a calcium/calmodulin (CaM)/calmodulin-dependent kinase II (CaMKII)-dependent manner similar to a mechanism whereby the N-methyl-D-aspartate (NMDA) receptor subunit NR2B facilitates chemical transmission. Since NR2B-microtubule interactions recruit receptors to the cell membrane during plasticity, we hypothesized an analogous modality for Cx36. We determined that Cx36 binding to tubulin at the carboxy-terminal domain was distinct from Cx43 and NR2B by binding a motif overlapping with the CaM and CaMKII binding motifs. Dual patch-clamp recordings demonstrated that pharmacological interference of the cytoskeleton and deleting the binding motif at the Cx36 carboxyl-terminal (CT) reversibly abolished Cx36 plasticity. Mechanistic details of trafficking to the gap-junction plaque (GJP) were probed pharmacologically and through mutational analysis, all of which affected GJP size and formation between cell pairs. Lys279, Ile280, and Lys281 positions were particularly critical. This study demonstrates that tubulin-dependent transport of Cx36 potentiates synaptic strength by delivering channels to GJPs, reinforcing the role of protein transport at chemical and electrical synapses to fine-tune communication between neurons.
Collapse
Affiliation(s)
- Cherie A Brown
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada.
| | - Cristiane Del Corsso
- Department of Biophysics and Physiology, Federal University of Rio de Janeiro-RJ, Rio de Janeiro 21941-901, Brazil.
| | - Christiane Zoidl
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada.
| | - Logan W Donaldson
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada.
| | - David C Spray
- Department of Neuroscience, Albert Einstein College, Bronx, NY 10461, USA.
- Department of Medicine, Albert Einstein College, Bronx, NY 10461, USA.
| | - Georg Zoidl
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada.
- Department of Psychology, York University, Toronto, ON M3J 1P3, Canada.
| |
Collapse
|
20
|
Yadav SC, Tetenborg S, Dedek K. Gap Junctions in A8 Amacrine Cells Are Made of Connexin36 but Are Differently Regulated Than Gap Junctions in AII Amacrine Cells. Front Mol Neurosci 2019; 12:99. [PMID: 31065239 PMCID: PMC6489437 DOI: 10.3389/fnmol.2019.00099] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 04/03/2019] [Indexed: 01/01/2023] Open
Abstract
In the mammalian retina, amacrine cells represent the most diverse cell class and are involved in the spatio-temporal processing of visual signals in the inner plexiform layer. They are connected to bipolar, other amacrine and ganglion cells, forming complex networks via electrical and chemical synapses. The small-field A8 amacrine cell was shown to receive non-selective glutamatergic input from OFF and ON cone bipolar cells at its bistratified dendrites in sublamina 1 and 4 of the inner plexiform layer. Interestingly, it was also shown to form electrical synapses with ON cone bipolar cells, thus resembling the rod pathway-specific AII amacrine cell. In contrast to the AII cell, however, the electrical synapses of A8 cells are poorly understood. Therefore, we made use of the Ier5-GFP mouse line, in which A8 cells are labeled by GFP, to study the gap junction composition and frequency in A8 cells. We found that A8 cells form <20 gap junctions per cell and these gap junctions consist of connexin36. Connexin36 is present at both OFF and ON dendrites of A8 cells, preferentially connecting A8 cells to type 1 OFF and type 6 and 7 ON bipolar cells and presumably other amacrine cells. Additionally, we show that the OFF dendrites of A8 cells co-stratify with the processes of dopaminergic amacrine cells from which they may receive GABAergic input via GABAA receptor subunit α3. As we found A8 cells to express dopamine receptor D1 (but not D2), we also tested whether A8 cell coupling is modulated by D1 receptor agonists and antagonists as was shown for the coupling of AII cells. However, this was not the case. In summary, our data suggests that A8 coupling is differently regulated than AII cells and may even be independent of ambient light levels and serve signal facilitation rather than providing a separate neuronal pathway.
Collapse
Affiliation(s)
- Shubhash C Yadav
- Animal Navigation/Neurosensorics, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany
| | - Stephan Tetenborg
- Animal Navigation/Neurosensorics, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany
| | - Karin Dedek
- Animal Navigation/Neurosensorics, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany.,Research Center Neurosensory Science, University of Oldenburg, Oldenburg, Germany
| |
Collapse
|
21
|
Farnsworth NL, Walter R, Piscopio RA, Schleicher WE, Benninger RKP. Exendin-4 overcomes cytokine-induced decreases in gap junction coupling via protein kinase A and Epac2 in mouse and human islets. J Physiol 2019; 597:431-447. [PMID: 30412665 PMCID: PMC6332825 DOI: 10.1113/jp276106] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 11/07/2018] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS The pancreatic islets of Langerhans maintain glucose homeostasis through insulin secretion, where insulin secretion dynamics are regulated by intracellular Ca2+ signalling and electrical coupling of the insulin producing β-cells in the islet. We have previously shown that cytokines decrease β-cell coupling and that compounds which increase cAMP can increase coupling. In both mouse and human islets exendin-4, which increases cAMP, protected against cytokine-induced decreases in coupling and in mouse islets preserved glucose-stimulated calcium signalling by increasing connexin36 gap junction levels on the plasma membrane. Our data indicate that protein kinase A regulates β-cell coupling through a fast mechanism, such as channel gating or membrane organization, while Epac2 regulates slower mechanisms of regulation, such as gap junction turnover. Increases in β-cell coupling with exendin-4 may protect against cytokine-mediated β-cell death as well as preserve insulin secretion dynamics during the development of diabetes. ABSTRACT The pancreatic islets of Langerhans maintain glucose homeostasis. Insulin secretion from islet β-cells is driven by glucose metabolism, depolarization of the cell membrane and an influx of calcium, which initiates the release of insulin. Gap junctions composed of connexin36 (Cx36) electrically couple β-cells, regulating calcium signalling and insulin secretion dynamics. Cx36 coupling is decreased in pre-diabetic mice, suggesting a role for altered coupling in diabetes. Our previous work has shown that pro-inflammatory cytokines decrease Cx36 coupling and that compounds which increase cAMP can increase Cx36 coupling. The goal of this study was to determine if exendin-4, which increases cAMP, can protect against cytokine-induced decreases in Cx36 coupling and altered islet function. In both mouse and human islets, exendin-4 protected against cytokine-induced decreases in coupling and preserved glucose-stimulated calcium signalling. Exendin-4 also protected against protein kinase Cδ-mediated decreases in Cx36 coupling. Exendin-4 preserved coupling in mouse islets by preserving Cx36 levels on the plasma membrane. Exendin-4 regulated Cx36 coupling via both protein kinase A (PKA)- and Epac2-mediated mechanisms in cytokine-treated islets. In mouse islets, modulating Epac2 had a greater impact in mediating Cx36 coupling, while in human islets modulating PKA had a greater impact on Cx36 coupling. Our data indicate that PKA regulates Cx36 coupling through a fast mechanism, such as channel gating, while Epac2 regulates slower mechanisms of regulation, such as Cx36 turnover in the membrane. Increases in Cx36 coupling with exendin-4 may protect against cytokine-mediated β-cell dysfunction to insulin secretion dynamics during the development of diabetes.
Collapse
Affiliation(s)
- Nikki L. Farnsworth
- Barbara Davis Center for Childhood DiabetesUniversity of Colorado Anschutz Medical CampusAuroraCO80045USA
| | - Rachelle Walter
- Department of BioengineeringUniversity of Colorado Anschutz Medical CampusAuroraCO80045USA
| | - Robert A. Piscopio
- Department of BioengineeringUniversity of Colorado Anschutz Medical CampusAuroraCO80045USA
| | - Wolfgang E. Schleicher
- Department of BioengineeringUniversity of Colorado Anschutz Medical CampusAuroraCO80045USA
| | - Richard K. P. Benninger
- Barbara Davis Center for Childhood DiabetesUniversity of Colorado Anschutz Medical CampusAuroraCO80045USA
- Department of BioengineeringUniversity of Colorado Anschutz Medical CampusAuroraCO80045USA
| |
Collapse
|
22
|
Lynn BD, Li X, Hormuzdi SG, Griffiths EK, McGlade CJ, Nagy JI. E3 ubiquitin ligases LNX1 and LNX2 localize at neuronal gap junctions formed by connexin36 in rodent brain and molecularly interact with connexin36. Eur J Neurosci 2018; 48:3062-3081. [PMID: 30295974 DOI: 10.1111/ejn.14198] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 08/31/2018] [Accepted: 09/25/2018] [Indexed: 12/31/2022]
Abstract
Electrical synapses in the mammalian central nervous system (CNS) are increasingly recognized as highly complex structures for mediation of neuronal communication, both with respect to their capacity for dynamic short- and long-term modification in efficacy of synaptic transmission and their multimolecular regulatory and structural components. These two characteristics are inextricably linked, such that understanding of mechanisms that contribute to electrical synaptic plasticity requires knowledge of the molecular composition of electrical synapses and the functions of proteins associated with these synapses. Here, we provide evidence that the key component of gap junctions that form the majority of electrical synapses in the mammalian CNS, namely connexin36 (Cx36), directly interacts with the related E3 ubiquitin ligase proteins Ligand of NUMB protein X1 (LNX1) and Ligand of NUMB protein X2 (LNX2). This is based on immunofluorescence colocalization of LNX1 and LNX2 with Cx36-containing gap junctions in adult mouse brain versus lack of such coassociation in LNX null mice, coimmunoprecipitation of LNX proteins with Cx36, and pull-down of Cx36 with the second PDZ domain of LNX1 and LNX2. Furthermore, cotransfection of cultured cells with Cx36 and E3 ubiquitin ligase-competent LNX1 and LNX2 isoforms led to loss of Cx36-containing gap junctions between cells, whereas these junctions persisted following transfection with isoforms of these proteins that lack ligase activity. Our results suggest that a LNX protein mediates ubiquitination of Cx36 at neuronal gap junctions, with consequent Cx36 internalization, and may thereby contribute to intracellular mechanisms that govern the recently identified modifiability of synaptic transmission at electrical synapses.
Collapse
Affiliation(s)
- Bruce D Lynn
- Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Xinbo Li
- Casey Eye Institute, Oregon Health and Science University, Portland, Oregon
| | - Sheriar G Hormuzdi
- D'Arcy Thompson Unit, School of Life Sciences, University of Dundee, Dundee, UK
| | - Emily K Griffiths
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - C Jane McGlade
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - James I Nagy
- Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Science, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
23
|
Welzel G, Schuster S. Long-term potentiation in an innexin-based electrical synapse. Sci Rep 2018; 8:12579. [PMID: 30135467 PMCID: PMC6105662 DOI: 10.1038/s41598-018-30966-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 08/09/2018] [Indexed: 11/08/2022] Open
Abstract
Electrical synapses are formed by two unrelated gap junction protein families, the primordial innexins (invertebrates) or the connexins (vertebrates). Although molecularly different, innexin- and connexin-based electrical synapses are strikingly similar in their membrane topology. However, it remains unclear if this similarity extends also to more sophisticated functions such as long-term potentiation which is only known in connexin-based synapses. Here we show that this capacity is not unique to connexin-based synapses. Using a method that allowed us to quantitatively measure gap-junction conductance we provide the first and unequivocal evidence of long-term potentiation in an innexin-based electrical synapse. Our findings suggest that long-term potentiation is a property that has likely existed already in ancestral gap junctions. They therefore could provide a highly potent system to dissect shared molecular mechanisms of electrical synapse plasticity.
Collapse
Affiliation(s)
- Georg Welzel
- Department of Animal Physiology, University of Bayreuth, 95440, Bayreuth, Germany.
| | - Stefan Schuster
- Department of Animal Physiology, University of Bayreuth, 95440, Bayreuth, Germany.
| |
Collapse
|
24
|
Nagy JI, Lynn BD. Structural and Intermolecular Associations Between Connexin36 and Protein Components of the Adherens Junction-Neuronal Gap Junction Complex. Neuroscience 2018; 384:241-261. [PMID: 29879437 DOI: 10.1016/j.neuroscience.2018.05.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/17/2018] [Accepted: 05/18/2018] [Indexed: 11/20/2022]
Abstract
Intimate structural and functional relationships between gap junctions and adherens junctions have been demonstrated in peripheral tissues, but have not been thoroughly examined in the central nervous system, where adherens junctions are often found in close proximity to neuronal gap junctions. Here, we used immunofluorescence approaches to document the localization of various protein components of adherens junctions in relation to those that we have previously reported to occur at electrical synapses formed by neuronal gap junctions composed of connexin36 (Cx36). The adherens junction constituents N-cadherin and nectin-1 were frequently found to localize near or overlap with Cx36-containing gap junctions in several brain regions examined. This was also true of the adherens junction-associated proteins α-catenin and β-catenin, as well as the proteins zonula occludens-1 and AF6 (aka, afadin) that were reported constituents of both adherens junctions and gap junctions. The deployment of the protein constituents of these junctions was especially striking at somatic contacts between primary afferent neurons in the mesencephalic trigeminal nucleus (MesV), where the structural components of adherens junctions appeared to be maintained in connexin36 null mice. These results support emerging views concerning the multi-molecular composition of electrical synapses and raise possibilities for various structural and functional protein-protein interactions at what now can be considered the adherens junction-neuronal gap junction complex. Further, the results point to intracellular signaling pathways that could potentially contribute to the assembly, maintenance and turnover of this complex, as well as to the dynamic nature of neuronal communication at electrical synapses.
Collapse
Affiliation(s)
- J I Nagy
- Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.
| | - B D Lynn
- Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
25
|
Nagy JI, Pereda AE, Rash JE. Electrical synapses in mammalian CNS: Past eras, present focus and future directions. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2018; 1860:102-123. [PMID: 28577972 PMCID: PMC5705454 DOI: 10.1016/j.bbamem.2017.05.019] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/26/2017] [Accepted: 05/27/2017] [Indexed: 12/19/2022]
Abstract
Gap junctions provide the basis for electrical synapses between neurons. Early studies in well-defined circuits in lower vertebrates laid the foundation for understanding various properties conferred by electrical synaptic transmission. Knowledge surrounding electrical synapses in mammalian systems unfolded first with evidence indicating the presence of gap junctions between neurons in various brain regions, but with little appreciation of their functional roles. Beginning at about the turn of this century, new approaches were applied to scrutinize electrical synapses, revealing the prevalence of neuronal gap junctions, the connexin protein composition of many of those junctions, and the myriad diverse neural systems in which they occur in the mammalian CNS. Subsequent progress indicated that electrical synapses constitute key elements in synaptic circuitry, govern the collective activity of ensembles of electrically coupled neurons, and in part orchestrate the synchronized neuronal network activity and rhythmic oscillations that underlie fundamental integrative processes. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- James I Nagy
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada.
| | - Alberto E Pereda
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, United States
| | - John E Rash
- Department of Biomedical Sciences, and Program in Molecular, Cellular and Integrative Neurosciences, Colorado State University, Fort Collins, CO 80523, United States
| |
Collapse
|
26
|
Bazzigaluppi P, Isenia SC, Haasdijk ED, Elgersma Y, De Zeeuw CI, van der Giessen RS, de Jeu MTG. Modulation of Murine Olivary Connexin 36 Gap Junctions by PKA and CaMKII. Front Cell Neurosci 2017; 11:397. [PMID: 29311830 PMCID: PMC5735106 DOI: 10.3389/fncel.2017.00397] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/28/2017] [Indexed: 11/15/2022] Open
Abstract
The inferior olive (IO) is a nucleus located in the brainstem and it is part of the olivo-cerebellar loop. This circuit plays a fundamental role in generation and acquisition of coherent motor patterns and it relies on synchronous activation of groups of Purkinje cells (PC) in the cerebellar cortex. IO neurons integrate their intrinsic oscillatory activity with excitatory inputs coming from the somatosensory system and inhibitory feedback coming from the cerebellar nuclei. Alongside these chemical synaptic inputs, IO neurons are coupled to one another via connexin 36 (Cx36) containing gap junctions (GJs) that create a functional syncytium between neurons. Communication between olivary neurons is regulated by these GJs and their correct functioning contributes to coherent oscillations in the IO and proper motor learning. Here, we explore the cellular pathways that can regulate the coupling between olivary neurons. We combined in vitro electrophysiology and immunohistochemistry (IHC) on mouse acute brain slices to unravel the pathways that regulate olivary coupling. We found that enhancing the activity of the protein kinase A (PKA) pathway and blocking the Ca2+/calmodulin-dependent protein kinase II (CaMKII) pathway can both down-regulate the size of the coupled network. However, these two kinases follow different mechanisms of action. Our results suggest that activation of the PKA pathway reduces the opening probability of the Cx36 GJs, whereas inhibition of the CaMKII pathway reduces the number of Cx36 GJs. The low densities of Cx36 proteins and electrical synapses in βCaMKII knock-out mice point towards an essential role for this protein kinase in regulating the density of GJs in the IO. Thus, the level of olivary coupling is a dynamic process and regulated by a variety of enzymes modulating GJs expression, docking and activity.
Collapse
Affiliation(s)
- Paolo Bazzigaluppi
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Sheena C Isenia
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Elize D Haasdijk
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Ype Elgersma
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | - Chris I De Zeeuw
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| | | | - Marcel T G de Jeu
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|
27
|
Leybaert L, Lampe PD, Dhein S, Kwak BR, Ferdinandy P, Beyer EC, Laird DW, Naus CC, Green CR, Schulz R. Connexins in Cardiovascular and Neurovascular Health and Disease: Pharmacological Implications. Pharmacol Rev 2017; 69:396-478. [PMID: 28931622 PMCID: PMC5612248 DOI: 10.1124/pr.115.012062] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Connexins are ubiquitous channel forming proteins that assemble as plasma membrane hemichannels and as intercellular gap junction channels that directly connect cells. In the heart, gap junction channels electrically connect myocytes and specialized conductive tissues to coordinate the atrial and ventricular contraction/relaxation cycles and pump function. In blood vessels, these channels facilitate long-distance endothelial cell communication, synchronize smooth muscle cell contraction, and support endothelial-smooth muscle cell communication. In the central nervous system they form cellular syncytia and coordinate neural function. Gap junction channels are normally open and hemichannels are normally closed, but pathologic conditions may restrict gap junction communication and promote hemichannel opening, thereby disturbing a delicate cellular communication balance. Until recently, most connexin-targeting agents exhibited little specificity and several off-target effects. Recent work with peptide-based approaches has demonstrated improved specificity and opened avenues for a more rational approach toward independently modulating the function of gap junctions and hemichannels. We here review the role of connexins and their channels in cardiovascular and neurovascular health and disease, focusing on crucial regulatory aspects and identification of potential targets to modify their function. We conclude that peptide-based investigations have raised several new opportunities for interfering with connexins and their channels that may soon allow preservation of gap junction communication, inhibition of hemichannel opening, and mitigation of inflammatory signaling.
Collapse
Affiliation(s)
- Luc Leybaert
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Paul D Lampe
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Stefan Dhein
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Brenda R Kwak
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Peter Ferdinandy
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Eric C Beyer
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Dale W Laird
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Christian C Naus
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Colin R Green
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| | - Rainer Schulz
- Physiology Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium (L.L.); Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington (P.D.L.); Institute for Pharmacology, University of Leipzig, Leipzig, Germany (S.D.); Department of Pathology and Immunology, Department of Medical Specialization-Cardiology, University of Geneva, Geneva, Switzerland (B.R.K.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Department of Pediatrics, University of Chicago, Chicago, Illinois (E.C.B.); Department of Anatomy and Cell Biology, University of Western Ontario, Dental Science Building, London, Ontario, Canada (D.W.L.); Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada (C.C.N.); Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand (C.R.G.); and Physiologisches Institut, Justus-Liebig-Universität Giessen, Giessen, Germany (R.S.)
| |
Collapse
|
28
|
Coulon P, Landisman CE. The Potential Role of Gap Junctional Plasticity in the Regulation of State. Neuron 2017; 93:1275-1295. [PMID: 28334604 DOI: 10.1016/j.neuron.2017.02.041] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 01/20/2017] [Accepted: 02/22/2017] [Indexed: 11/19/2022]
Abstract
Electrical synapses are the functional correlate of gap junctions and allow transmission of small molecules and electrical current between coupled neurons. Instead of static pores, electrical synapses are actually plastic, similar to chemical synapses. In the thalamocortical system, gap junctions couple inhibitory neurons that are similar in their biochemical profile, morphology, and electrophysiological properties. We postulate that electrical synaptic plasticity among inhibitory neurons directly interacts with the switching between different firing patterns in a state-dependent and type-dependent manner. In neuronal networks, electrical synapses may function as a modifiable resonance feedback system that enables stable oscillations. Furthermore, the plasticity of electrical synapses may play an important role in regulation of state, synchrony, and rhythmogenesis in the mammalian thalamocortical system, similar to chemical synaptic plasticity. Based on their plasticity, rich diversity, and specificity, electrical synapses are thus likely to participate in the control of consciousness and attention.
Collapse
Affiliation(s)
- Philippe Coulon
- Seattle Children's Research Institute, Center for Integrative Brain Research, Seattle, WA 98101, USA.
| | - Carole E Landisman
- Seattle Children's Research Institute, Center for Integrative Brain Research, Seattle, WA 98101, USA.
| |
Collapse
|
29
|
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: 0.6] [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
|
30
|
Westacott MJ, Farnsworth NL, St Clair JR, Poffenberger G, Heintz A, Ludin NW, Hart NJ, Powers AC, Benninger RKP. Age-Dependent Decline in the Coordinated [Ca 2+] and Insulin Secretory Dynamics in Human Pancreatic Islets. Diabetes 2017; 66:2436-2445. [PMID: 28588099 PMCID: PMC5566297 DOI: 10.2337/db17-0137] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 05/30/2017] [Indexed: 12/25/2022]
Abstract
Aging is associated with increased risk for type 2 diabetes, resulting from reduced insulin sensitivity and secretion. Reduced insulin secretion can result from reduced proliferative capacity and reduced islet function. Mechanisms underlying altered β-cell function in aging are poorly understood in mouse and human islets, and the impact of aging on intraislet communication has not been characterized. Here, we examine how β-cell [Ca2+] and electrical communication are impacted during aging in mouse and human islets. Islets from human donors and from mice were studied using [Ca2+] imaging, static and perifusion insulin secretion assays, and gap junction permeability measurements. In human islets, [Ca2+] dynamics were coordinated within distinct subregions of the islet, invariant with islet size. There was a marked decline in the coordination of [Ca2+] dynamics, gap junction coupling, and insulin secretion dynamics with age. These age-dependent declines were reversed by pharmacological gap junction activation. These results show that human islet function declines with aging, which can reduce insulin action and may contribute to increased risk of type 2 diabetes.
Collapse
Affiliation(s)
- Matthew J Westacott
- Department of Bioengineering, University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Nikki L Farnsworth
- Barbara Davis Center for Diabetes, University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Joshua R St Clair
- Department of Bioengineering, University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Greg Poffenberger
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN
- VA Tennessee Valley Healthcare System, Nashville, TN
| | - Audrey Heintz
- Department of Bioengineering, University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Nurin W Ludin
- Department of Bioengineering, University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Nathaniel J Hart
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Alvin C Powers
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN
- VA Tennessee Valley Healthcare System, Nashville, TN
| | - Richard K P Benninger
- Department of Bioengineering, University of Colorado, Anschutz Medical Campus, Aurora, CO
- Barbara Davis Center for Diabetes, University of Colorado, Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
31
|
Roy S, Kim D, Lim R. Cell-cell communication in diabetic retinopathy. Vision Res 2017; 139:115-122. [PMID: 28583293 DOI: 10.1016/j.visres.2017.04.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/27/2017] [Accepted: 04/28/2017] [Indexed: 10/19/2022]
Abstract
In diabetic retinopathy, high glucose (HG)-mediated breakdown in cell-cell communication promotes disruption of retinal homeostasis. Several studies indicate that HG condition alters expression of connexin genes and subsequent gap junction intercellular communication (GJIC) in retinal vascular cells and non-vascular cells. A serious consequence of disrupted cell-cell communication is apoptosis and breakdown of the blood-retinal barrier (BRB). More recently, studies suggest adverse effects from HG on retinal Müller cells. This article focuses on HG-mediated changes in connexin expression and GJIC and their subsequent effects on the breakdown of retinal homeostasis, cell death, compromised vascular permeability, and interactions between endothelial cells, pericytes and retinal Müller cells in the pathogenesis of diabetic retinopathy. Additionally, options for rectifying disrupted homeostasis under HG condition associated with diabetic retinopathy are reviewed.
Collapse
Affiliation(s)
- Sayon Roy
- Department of Medicine and Ophthalmology, Boston University School of Medicine, Boston, MA, United States.
| | - Dongjoon Kim
- Department of Medicine and Ophthalmology, Boston University School of Medicine, Boston, MA, United States
| | - Remington Lim
- Department of Medicine and Ophthalmology, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
32
|
Pérez de Sevilla Müller L, Solomon A, Sheets K, Hapukino H, Rodriguez AR, Brecha NC. Multiple cell types form the VIP amacrine cell population. J Comp Neurol 2017; 527:133-158. [PMID: 28472856 DOI: 10.1002/cne.24234] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 04/21/2017] [Accepted: 04/27/2017] [Indexed: 12/21/2022]
Abstract
Amacrine cells are a heterogeneous group of interneurons that form microcircuits with bipolar, amacrine and ganglion cells to process visual information in the inner retina. This study has characterized the morphology, neurochemistry and major cell types of a VIP-ires-Cre amacrine cell population. VIP-tdTomato and -Confetti (Brainbow2.1) mouse lines were generated by crossing a VIP-ires-Cre line with either a Cre-dependent tdTomato or Brainbow2.1 reporter line. Retinal sections and whole-mounts were evaluated by quantitative, immunohistochemical, and intracellular labeling approaches. The majority of tdTomato and Confetti fluorescent cell bodies were in the inner nuclear layer (INL) and a few cell bodies were in the ganglion cell layer (GCL). Fluorescent processes ramified in strata 1, 3, 4, and 5 of the inner plexiform layer (IPL). All tdTomato fluorescent cells expressed syntaxin 1A and GABA-immunoreactivity indicating they were amacrine cells. The average VIP-tdTomato fluorescent cell density in the INL and GCL was 535 and 24 cells/mm2 , respectively. TdTomato fluorescent cells in the INL and GCL contained VIP-immunoreactivity. The VIP-ires-Cre amacrine cell types were identified in VIP-Brainbow2.1 retinas or by intracellular labeling in VIP-tdTomato retinas. VIP-1 amacrine cells are bistratified, wide-field cells that ramify in strata 1, 4, and 5, VIP-2A and 2B amacrine cells are medium-field cells that mainly ramify in strata 3 and 4, and VIP-3 displaced amacrine cells are medium-field cells that ramify in strata 4 and 5 of the IPL. VIP-ires-Cre amacrine cells form a neuropeptide-expressing cell population with multiple cell types, which are likely to have distinct roles in visual processing.
Collapse
Affiliation(s)
- Luis Pérez de Sevilla Müller
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763
| | - Alexander Solomon
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763
| | - Kristopher Sheets
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763
| | - Hinekura Hapukino
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763
| | - Allen R Rodriguez
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763
| | - Nicholas C Brecha
- Department of Neurobiology, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763.,Department of Medicine, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763.,Department of Ophthalmology and the Stein Eye Institute, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763.,CURE Digestive Diseases Research Center, David Geffen School of Medicine at Los Angeles, University of California at Los Angeles, Los Angeles, California, 90095-1763.,Veterans Administration Greater Los Angeles Health System, Los Angeles, California, 90073
| |
Collapse
|
33
|
Pérez de Sevilla Müller L, Azar SS, de Los Santos J, Brecha NC. Prox1 Is a Marker for AII Amacrine Cells in the Mouse Retina. Front Neuroanat 2017; 11:39. [PMID: 28529477 PMCID: PMC5418924 DOI: 10.3389/fnana.2017.00039] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 04/18/2017] [Indexed: 12/18/2022] Open
Abstract
The transcription factor Prox1 is expressed in multiple cells in the retina during eye development. This study has focused on neuronal Prox1 expression in the inner nuclear layer (INL) of the adult mouse retina. Prox1 immunostaining was evaluated in vertical retinal sections and whole mount preparations using a specific antibody directed to the C-terminus of Prox1. Strong immunostaining was observed in numerous amacrine cell bodies and in all horizontal cell bodies in the proximal and distal INL, respectively. Some bipolar cells were also weakly immunostained. Prox1-immunoreactive amacrine cells expressed glycine, and they formed 35 ± 3% of all glycinergic amacrine cells. Intracellular Neurobiotin injections into AII amacrine cells showed that all gap junction-coupled AII amacrine cells express Prox1, and no other Prox1-immunostained amacrine cells were in the immediate area surrounding the injected AII amacrine cell. Prox1-immunoreactive amacrine cell bodies were distributed across the retina, with their highest density (3887 ± 160 cells/mm2) in the central retina, 0.5 mm from the optic nerve head, and their lowest density (3133 ± 350 cells/mm2) in the mid-peripheral retina, 2 mm from the optic nerve head. Prox1-immunoreactive amacrine cell bodies comprised ~9.8% of the total amacrine cell population, and they formed a non-random mosaic with a regularity index (RI) of 3.4, similar to AII amacrine cells in the retinas of other mammals. Together, these findings indicate that AII amacrine cells are the predominant and likely only amacrine cell type strongly expressing Prox1 in the adult mouse retina, and establish Prox1 as a marker of AII amacrine cells.
Collapse
Affiliation(s)
- Luis Pérez de Sevilla Müller
- Departments of Neurobiology, Medicine and Ophthalmology, David Geffen School of Medicine at Los Angeles, University of California, Los AngelesLos Angeles, CA, USA
| | - Shaghauyegh S Azar
- Departments of Neurobiology, Medicine and Ophthalmology, David Geffen School of Medicine at Los Angeles, University of California, Los AngelesLos Angeles, CA, USA
| | - Janira de Los Santos
- Departments of Neurobiology, Medicine and Ophthalmology, David Geffen School of Medicine at Los Angeles, University of California, Los AngelesLos Angeles, CA, USA
| | - Nicholas C Brecha
- Departments of Neurobiology, Medicine and Ophthalmology, David Geffen School of Medicine at Los Angeles, University of California, Los AngelesLos Angeles, CA, USA.,Stein Eye Institute, David Geffen School of Medicine at Los Angeles, University of California, Los AngelesLos Angeles, CA, USA.,CURE Digestive Diseases Research Center, David Geffen School of Medicine at Los Angeles, University of California, Los AngelesLos Angeles, CA, USA.,Veterans Administration Greater Los Angeles Health SystemLos Angeles, CA, USA
| |
Collapse
|
34
|
Mathews J, Levin M. Gap junctional signaling in pattern regulation: Physiological network connectivity instructs growth and form. Dev Neurobiol 2017; 77:643-673. [PMID: 27265625 PMCID: PMC10478170 DOI: 10.1002/dneu.22405] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 05/27/2016] [Accepted: 05/31/2016] [Indexed: 12/19/2022]
Abstract
Gap junctions (GJs) are aqueous channels that allow cells to communicate via physiological signals directly. The role of gap junctional connectivity in determining single-cell functions has long been recognized. However, GJs have another important role: the regulation of large-scale anatomical pattern. GJs are not only versatile computational elements that allow cells to control which small molecule signals they receive and emit, but also establish connectivity patterns within large groups of cells. By dynamically regulating the topology of bioelectric networks in vivo, GJs underlie the ability of many tissues to implement complex morphogenesis. Here, a review of recent data on patterning roles of GJs in growth of the zebrafish fin, the establishment of left-right patterning, the developmental dysregulation known as cancer, and the control of large-scale head-tail polarity, and head shape in planarian regeneration has been reported. A perspective in which GJs are not only molecular features functioning in single cells, but also enable global neural-like dynamics in non-neural somatic tissues has been proposed. This view suggests a rich program of future work which capitalizes on the rapid advances in the biophysics of GJs to exploit GJ-mediated global dynamics for applications in birth defects, regenerative medicine, and morphogenetic bioengineering. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 643-673, 2017.
Collapse
Affiliation(s)
- Juanita Mathews
- Department of Biology, Tufts Center for Regenerative and Developmental Biology, Tufts University, Medford, MA
| | - Michael Levin
- Department of Biology, Tufts Center for Regenerative and Developmental Biology, Tufts University, Medford, MA
| |
Collapse
|
35
|
Miller AC, Pereda AE. The electrical synapse: Molecular complexities at the gap and beyond. Dev Neurobiol 2017; 77:562-574. [PMID: 28170151 DOI: 10.1002/dneu.22484] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/04/2017] [Accepted: 01/04/2017] [Indexed: 12/21/2022]
Abstract
Gap junctions underlie electrical synaptic transmission between neurons. Generally perceived as simple intercellular channels, "electrical synapses" have demonstrated to be more functionally sophisticated and structurally complex than initially anticipated. Electrical synapses represent an assembly of multiple molecules, consisting of channels, adhesion complexes, scaffolds, regulatory machinery, and trafficking proteins, all required for their proper function and plasticity. Additionally, while electrical synapses are often viewed as strictly symmetric structures, emerging evidence has shown that some components forming electrical synapses can be differentially distributed at each side of the junction. We propose that the molecular complexity and asymmetric distribution of proteins at the electrical synapse provides rich potential for functional diversity. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 562-574, 2017.
Collapse
Affiliation(s)
- Adam C Miller
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon
| | - Alberto E Pereda
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
36
|
Belousov AB, Fontes JD, Freitas-Andrade M, Naus CC. Gap junctions and hemichannels: communicating cell death in neurodevelopment and disease. BMC Cell Biol 2017; 18:4. [PMID: 28124625 PMCID: PMC5267333 DOI: 10.1186/s12860-016-0120-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Gap junctions are unique membrane channels that play a significant role in intercellular communication in the developing and mature central nervous system (CNS). These channels are composed of connexin proteins that oligomerize into hexamers to form connexons or hemichannels. Many different connexins are expressed in the CNS, with some specificity with regard to the cell types in which distinct connexins are found, as well as the timepoints when they are expressed in the developing and mature CNS. Both the main neuronal Cx36 and glial Cx43 play critical roles in neurodevelopment. These connexins also mediate distinct aspects of the CNS response to pathological conditions. An imbalance in the expression, translation, trafficking and turnover of connexins, as well as mutations of connexins, can impact their function in the context of cell death in neurodevelopment and disease. With the ever-increasing understanding of connexins in the brain, therapeutic strategies could be developed to target these membrane channels in various neurological disorders.
Collapse
Affiliation(s)
- Andrei B Belousov
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, The University of Kansas, Kansas City, KS, 66160, USA
| | - Joseph D Fontes
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, The University of Kansas, Kansas City, KS, 66160, USA
| | - Moises Freitas-Andrade
- Department of Cellular & Physiological Sciences, Faculty of Medicine, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Christian C Naus
- Department of Cellular & Physiological Sciences, Faculty of Medicine, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
37
|
Sullivan KG, Emmons-Bell M, Levin M. Physiological inputs regulate species-specific anatomy during embryogenesis and regeneration. Commun Integr Biol 2016; 9:e1192733. [PMID: 27574538 PMCID: PMC4988443 DOI: 10.1080/19420889.2016.1192733] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 05/13/2016] [Accepted: 05/16/2016] [Indexed: 12/12/2022] Open
Abstract
A key problem in evolutionary developmental biology is identifying the sources of instructive information that determine species-specific anatomical pattern. Understanding the inputs to large-scale morphology is also crucial for efforts to manipulate pattern formation in regenerative medicine and synthetic bioengineering. Recent studies have revealed a physiological system of communication among cells that regulates pattern during embryogenesis and regeneration in vertebrate and invertebrate models. Somatic tissues form networks using the same ion channels, electrical synapses, and neurotransmitter mechanisms exploited by the brain for information-processing. Experimental manipulation of these circuits was recently shown to override genome default patterning outcomes, resulting in head shapes resembling those of other species in planaria and Xenopus. The ability to drastically alter macroscopic anatomy to that of other extant species, despite a wild-type genomic sequence, suggests exciting new approaches to the understanding and control of patterning. Here, we review these results and discuss hypotheses regarding non-genomic systems of instructive information that determine biological growth and form.
Collapse
Affiliation(s)
- Kelly G Sullivan
- Allen Discovery Center at Tufts University, Tufts University , Medford, MA, USA
| | - Maya Emmons-Bell
- Allen Discovery Center at Tufts University, Tufts University , Medford, MA, USA
| | - Michael Levin
- Allen Discovery Center at Tufts University, Tufts University , Medford, MA, USA
| |
Collapse
|
38
|
Kohmann D, Lüttjohann A, Seidenbecher T, Coulon P, Pape HC. Short-term depression of gap junctional coupling in reticular thalamic neurons of absence epileptic rats. J Physiol 2016; 594:5695-710. [PMID: 26940972 DOI: 10.1113/jp271811] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 03/02/2016] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Gap junctional electrical coupling between neurons of the reticular thalamic nucleus (RTN) is critical for hypersynchrony in the thalamo-cortical network. This study investigates the role of electrical coupling in pathological rhythmogenesis in RTN neurons in a rat model of absence epilepsy. Rhythmic activation resulted in a Ca(2+) -dependent short-term depression (STD) of electrical coupling between pairs of RTN neurons in epileptic rats, but not in RTN of a non-epileptic control strain. Pharmacological blockade of gap junctions in RTN in vivo induced a depression of seizure activity. The STD of electrical coupling represents a mechanism of Ca(2+) homeostasis in RTN aimed to counteract excessive synchronization. ABSTRACT Neurons in the reticular thalamic nucleus (RTN) are coupled by electrical synapses, which play a major role in regulating synchronous activity. This study investigates electrical coupling in RTN neurons from a rat model of childhood absence epilepsy, genetic absence epilepsy rats from Strasbourg (GAERS), compared with a non-epileptic control (NEC) strain, to assess the impact on pathophysiological rhythmogenesis. Whole-cell recordings were obtained from pairs of RTN neurons of GAERS and NEC in vitro. Coupling was determined by injection of hyperpolarizing current steps in one cell and monitoring evoked voltage responses in both activated and coupled cell. The coupling coefficient (cc) was compared under resting condition, during pharmacological interventions and repeated activation using a series of current injections. The effect of gap junctional coupling on seizure expression was investigated by application of gap junctional blockers into RTN of GAERS in vivo. At resting conditions, cc did not differ between GAERS and NEC. During repeated activation, cc declined in GAERS but not in NEC. This depression in cc was restored within 25 s and was prevented by intracellular presence of BAPTA in the activated but not in the coupled cell. Local application of gap junctional blockers into RTN of GAERS in vivo resulted in a decrease of spike wave discharge (SWD) activity. Repeated activation results in a short-term depression (STD) of gap junctional coupling in RTN neurons of GAERS, depending on intracellular Ca(2+) mechanisms in the activated cell. As blockage of gap junctions in vivo results in a decrease of SWD activity, the STD observed in GAERS is considered a compensatory mechanism, aimed to dampen SWD activity.
Collapse
Affiliation(s)
| | | | - Thomas Seidenbecher
- Institute of Physiology I, Westfälische Wilhelms-University Münster, Münster, Germany
| | | | | |
Collapse
|
39
|
Pogoda K, Kameritsch P, Retamal MA, Vega JL. Regulation of gap junction channels and hemichannels by phosphorylation and redox changes: a revision. BMC Cell Biol 2016; 17 Suppl 1:11. [PMID: 27229925 PMCID: PMC4896245 DOI: 10.1186/s12860-016-0099-3] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Post-translational modifications of connexins play an important role in the regulation of gap junction and hemichannel permeability. The prerequisite for the formation of functional gap junction channels is the assembly of connexin proteins into hemichannels and their insertion into the membrane. Hemichannels can affect cellular processes by enabling the passage of signaling molecules between the intracellular and extracellular space. For the intercellular communication hemichannels from one cell have to dock to its counterparts on the opposing membrane of an adjacent cell to allow the transmission of signals via gap junctions from one cell to the other. The controlled opening of hemichannels and gating properties of complete gap junctions can be regulated via post-translational modifications of connexins. Not only channel gating, but also connexin trafficking and assembly into hemichannels can be affected by post-translational changes. Recent investigations have shown that connexins can be modified by phosphorylation/dephosphorylation, redox-related changes including effects of nitric oxide (NO), hydrogen sulfide (H2S) or carbon monoxide (CO), acetylation, methylation or ubiquitination. Most of the connexin isoforms are known to be phosphorylated, e.g. Cx43, one of the most studied connexin at all, has 21 reported phosphorylation sites. In this review, we provide an overview about the current knowledge and relevant research of responsible kinases, connexin phosphorylation sites and reported effects on gap junction and hemichannel regulation. Regarding the effects of oxidants we discuss the role of NO in different cell types and tissues and recent studies about modifications of connexins by CO and H2S.
Collapse
Affiliation(s)
- Kristin Pogoda
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München and Munich University Hospital, München, Germany. .,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, München, Germany.
| | - Petra Kameritsch
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München and Munich University Hospital, München, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, München, Germany
| | - Mauricio A Retamal
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - José L Vega
- Experimental Physiology Laboratory (EPhyL), Antofagasta Institute, Universidad de Antofagasta, Antofagasta, Chile
| |
Collapse
|
40
|
Tu HY, Chiao CC. Cx36 expression in the AII-mediated rod pathway is activity dependent in the developing rabbit retina. Dev Neurobiol 2016; 76:473-86. [PMID: 26084632 DOI: 10.1002/dneu.22320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 06/09/2015] [Accepted: 06/11/2015] [Indexed: 12/20/2022]
Abstract
Gap junctions are composed of connexin 36 (Cx36) and play a critical role in the rod photoreceptor signaling pathways of the vertebrate retina. Despite the fact that their connection and modulation in various rod pathways have been extensively studied in adult animals, little is known about the contribution and regulation of gap junctions to the development of the AII amacrine cell (AC)-mediated rod pathway. Using immunohistochemistry and microinjection, this study demonstrates a steady increase in relative Cx36 protein expression in both plexiform layers of the rabbit retina at around the time of eye opening. However, immediately after eye opening, most Cx36 immunoreactive AII ACs show no gap junction coupling pattern to neighboring cells and it is not until the third postnatal week that AII cells begin to exhibit an adult-like tracer-coupling pattern. Moreover, studies using dark-rearing and AMPA receptor blockade during postnatal development both revealed that relative levels of Cx36 immunoreactivity in AII ACs were increased when neural activity was inhibited. Our findings suggest that Cx36 expression in the AII-mediated rod pathway is activity dependent in the developing rabbit retina.
Collapse
Affiliation(s)
- Hung-Ya Tu
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan
- Department of Life Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chuan-Chin Chiao
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan
- Department of Life Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, 30013, Taiwan
| |
Collapse
|
41
|
Farnsworth NL, Walter RL, Hemmati A, Westacott MJ, Benninger RKP. Low Level Pro-inflammatory Cytokines Decrease Connexin36 Gap Junction Coupling in Mouse and Human Islets through Nitric Oxide-mediated Protein Kinase Cδ. J Biol Chem 2016; 291:3184-96. [PMID: 26668311 PMCID: PMC4751367 DOI: 10.1074/jbc.m115.679506] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 12/07/2015] [Indexed: 11/06/2022] Open
Abstract
Pro-inflammatory cytokines contribute to the decline in islet function during the development of diabetes. Cytokines can disrupt insulin secretion and calcium dynamics; however, the mechanisms underlying this are poorly understood. Connexin36 gap junctions coordinate glucose-induced calcium oscillations and pulsatile insulin secretion across the islet. Loss of gap junction coupling disrupts these dynamics, similar to that observed during the development of diabetes. This study investigates the mechanisms by which pro-inflammatory cytokines mediate gap junction coupling. Specifically, as cytokine-induced NO can activate PKCδ, we aimed to understand the role of PKCδ in modulating cytokine-induced changes in gap junction coupling. Isolated mouse and human islets were treated with varying levels of a cytokine mixture containing TNF-α, IL-1β, and IFN-γ. Islet dysfunction was measured by insulin secretion, calcium dynamics, and gap junction coupling. Modulators of PKCδ and NO were applied to determine their respective roles in modulating gap junction coupling. High levels of cytokines caused cell death and decreased insulin secretion. Low levels of cytokine treatment disrupted calcium dynamics and decreased gap junction coupling, in the absence of disruptions to insulin secretion. Decreases in gap junction coupling were dependent on NO-regulated PKCδ, and altered membrane organization of connexin36. This study defines several mechanisms underlying the disruption to gap junction coupling under conditions associated with the development of diabetes. These mechanisms will allow for greater understanding of islet dysfunction and suggest ways to ameliorate this dysfunction during the development of diabetes.
Collapse
Affiliation(s)
- Nikki L Farnsworth
- From the Barbara Davis Center for Childhood Diabetes, Department of Bioengineering, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045
| | - Rachelle L Walter
- Department of Bioengineering, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045
| | - Alireza Hemmati
- Department of Bioengineering, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045
| | - Matthew J Westacott
- Department of Bioengineering, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045
| | - Richard K P Benninger
- From the Barbara Davis Center for Childhood Diabetes, Department of Bioengineering, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045
| |
Collapse
|
42
|
Farshi P, Fyk-Kolodziej B, Krolewski DM, Walker PD, Ichinose T. Dopamine D1 receptor expression is bipolar cell type-specific in the mouse retina. J Comp Neurol 2015; 524:2059-79. [PMID: 26587737 DOI: 10.1002/cne.23932] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 11/16/2015] [Accepted: 11/17/2015] [Indexed: 01/25/2023]
Abstract
In the retina, dopamine is a key molecule for daytime vision. Dopamine is released by retinal dopaminergic amacrine cells and transmits signaling either by conventional synaptic or by volume transmission. By means of volume transmission, dopamine modulates all layers of retinal neurons; however, it is not well understood how dopamine modulates visual signaling pathways in bipolar cells. Here we analyzed Drd1a-tdTomato BAC transgenic mice and found that the dopamine D1 receptor (D1R) is expressed in retinal bipolar cells in a type-dependent manner. Strong tdTomato fluorescence was detected in the inner nuclear layer and localized to type 1, 3b, and 4 OFF bipolar cells and type 5-2, XBC, 6, and 7 ON bipolar cells. In contrast, type 2, 3a, 5-1, 9, and rod bipolar cells did not express Drd1a-tdTomato. Other interneurons were also found to express tdTomato including horizontal cells and a subset (25%) of AII amacrine cells. Diverse visual processing pathways, such as color or motion-coded pathways, are thought to be initiated in retinal bipolar cells. Our results indicate that dopamine sculpts bipolar cell performance in a type-dependent manner to facilitate daytime vision. J. Comp. Neurol. 524:2059-2079, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Pershang Farshi
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Bozena Fyk-Kolodziej
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - David M Krolewski
- Molecular and Behavioral Neuroscience Institute, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Paul D Walker
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Tomomi Ichinose
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Ophthalmology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
43
|
Activation of Group I and Group II Metabotropic Glutamate Receptors Causes LTD and LTP of Electrical Synapses in the Rat Thalamic Reticular Nucleus. J Neurosci 2015; 35:7616-25. [PMID: 25972185 DOI: 10.1523/jneurosci.3688-14.2015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Compared with the extensive characterization of chemical synaptic plasticity, electrical synaptic plasticity remains poorly understood. Electrical synapses are strong and prevalent among the GABAergic neurons of the rodent thalamic reticular nucleus. Using paired whole-cell recordings, we show that activation of Group I metabotropic glutamate receptors (mGluRs) induces long-term depression of electrical synapses. Conversely, activation of the Group II mGluR, mGluR3, induces long-term potentiation of electrical synapses. By testing downstream targets, we show that modifications induced by both mGluR groups converge on the same signaling cascade--adenylyl cyclase to cAMP to protein kinase A--but with opposing effects. Furthermore, the magnitude of modification is inversely correlated to baseline coupling strength. Thus, electrical synapses, like their chemical counterparts, undergo both strengthening and weakening forms of plasticity, which should play a significant role in thalamocortical function.
Collapse
|
44
|
Nakamura Y, Morioka N, Zhang FF, Hisaoka-Nakashima K, Nakata Y. Downregulation of connexin36 in mouse spinal dorsal horn neurons leads to mechanical allodynia. J Neurosci Res 2015; 93:584-91. [PMID: 25400139 DOI: 10.1002/jnr.23515] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 09/24/2014] [Accepted: 10/13/2014] [Indexed: 11/08/2022]
Abstract
Connexin36 (Cx36), a component of neuronal gap junctions, is crucial for interneuronal communication and regulation. Gap junction dysfunction underlies neurological disorders, including chronic pain. Following a peripheral nerve injury, Cx36 expression in the ipsilateral spinal dorsal horn was markedly decreased over time, which paralleled the time course of hind paw tactile allodynia. Intrathecal (i.t.) injection of Cx36 siRNA (1 and 5 pg) significantly reduced the expression of Cx36 protein in the lumbar spinal cord, peaking 3 days after the injection, which corresponded with the onset of hind paw tactile allodynia. It is possible that some of the tactile allodynia resulting from Cx36 downregulation could be mediated through excitatory neuromodulators, such as glutamate and substance P. The Cx36 knockdown-evoked tactile allodynia was significantly attenuated by i.t. treatment with the N-methyl-D-aspartate glutamate receptor antagonist MK-801 but not the substance P receptor antagonist CP96345. Immunohistochemistry showed that Cx36 was colocalized with glycine transporter-2, a marker for inhibitory glycinergic spinal interneurons, but not with glutamate decarboxylase 67, a marker for inhibitory GABAergic spinal interneurons. The results indicate that spinal inhibition through glycinergic interneurons is reduced, leading to increased glutamatergic neurotransmission, as a result of Cx36 downregulation. The current data suggest that gap junction dysfunction underlies neuropathic pain and further suggest a novel target for the development of analgesics.
Collapse
Affiliation(s)
- Yoki Nakamura
- Department of Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | | | | | | | | |
Collapse
|
45
|
Benninger RKP, Hutchens T, Head WS, McCaughey MJ, Zhang M, Le Marchand SJ, Satin LS, Piston DW. Intrinsic islet heterogeneity and gap junction coupling determine spatiotemporal Ca²⁺ wave dynamics. Biophys J 2014; 107:2723-33. [PMID: 25468351 DOI: 10.1016/j.bpj.2014.10.048] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 09/09/2014] [Accepted: 10/08/2014] [Indexed: 10/24/2022] Open
Abstract
Insulin is released from the islets of Langerhans in discrete pulses that are linked to synchronized oscillations of intracellular free calcium ([Ca(2+)]i). Associated with each synchronized oscillation is a propagating calcium wave mediated by Connexin36 (Cx36) gap junctions. A computational islet model predicted that waves emerge due to heterogeneity in β-cell function throughout the islet. To test this, we applied defined patterns of glucose stimulation across the islet using a microfluidic device and measured how these perturbations affect calcium wave propagation. We further investigated how gap junction coupling regulates spatiotemporal [Ca(2+)]i dynamics in the face of heterogeneous glucose stimulation. Calcium waves were found to originate in regions of the islet having elevated excitability, and this heterogeneity is an intrinsic property of islet β-cells. The extent of [Ca(2+)]i elevation across the islet in the presence of heterogeneity is gap-junction dependent, which reveals a glucose dependence of gap junction coupling. To better describe these observations, we had to modify the computational islet model to consider the electrochemical gradient between neighboring β-cells. These results reveal how the spatiotemporal [Ca(2+)]i dynamics of the islet depend on β-cell heterogeneity and cell-cell coupling, and are important for understanding the regulation of coordinated insulin release across the islet.
Collapse
Affiliation(s)
- Richard K P Benninger
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee; Department of Bioengineering, University of Colorado, Aurora, Colorado; Barbara Davis Center, University of Colorado, Aurora, Colorado.
| | - Troy Hutchens
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee
| | - W Steven Head
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee
| | - Michael J McCaughey
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee
| | - Min Zhang
- Department of Pharmacology, Virginia Commonwealth University, Richmond, Virginia
| | - Sylvain J Le Marchand
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee
| | - Leslie S Satin
- Department of Pharmacology, Virginia Commonwealth University, Richmond, Virginia; Department of Pharmacology, University of Michigan, Ann Arbor, Michigan; Brehm Diabetes Center, University of Michigan, Ann Arbor, Michigan
| | - David W Piston
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee.
| |
Collapse
|
46
|
Tian N, Xu HP, Wang P. Dopamine D2 receptors preferentially regulate the development of light responses of the inner retina. Eur J Neurosci 2014; 41:17-30. [PMID: 25393815 DOI: 10.1111/ejn.12783] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 09/25/2014] [Accepted: 10/13/2014] [Indexed: 01/14/2023]
Abstract
Retinal light responsiveness measured via electroretinography undergoes developmental modulation, and is thought to be critically regulated by both visual experience and dopamine. The primary goal of this study was to determine whether dopamine D2 receptors regulate the visual experience-dependent functional development of the retina. Accordingly, we recorded electroretinograms from wild-type mice and mice with a genetic deletion of the gene that encodes the D2 receptor raised under normal cyclic light conditions and constant darkness. Our results demonstrate that D2 receptor mutation preferentially increases the amplitude of the inner retinal light responses evoked by high-intensity light measured as oscillatory potentials in adult mice. During postnatal development, all three major components of electroretinograms, i.e. a-waves, b-waves, and oscillatory potentials, increase with age. Comparatively, D2 receptor mutation preferentially reduces the age-dependent increase in b-waves evoked by low-intensity light. Light deprivation from birth reduces b-wave amplitudes and completely abolishes the increased amplitude of oscillatory potentials of D2 receptor mutants. Taken together, these results demonstrate that D2 receptors play an important role in the activity-dependent functional development of the mouse retina.
Collapse
Affiliation(s)
- Ning Tian
- Department of Ophthalmology and Visual Science, University of Utah School of Medicine, Salt Lake City, UT, 84132, USA
| | | | | |
Collapse
|
47
|
Electrical synapses and their functional interactions with chemical synapses. Nat Rev Neurosci 2014; 15:250-63. [PMID: 24619342 DOI: 10.1038/nrn3708] [Citation(s) in RCA: 326] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Brain function relies on the ability of neurons to communicate with each other. Interneuronal communication primarily takes place at synapses, where information from one neuron is rapidly conveyed to a second neuron. There are two main modalities of synaptic transmission: chemical and electrical. Far from functioning independently and serving unrelated functions, mounting evidence indicates that these two modalities of synaptic transmission closely interact, both during development and in the adult brain. Rather than conceiving synaptic transmission as either chemical or electrical, this article emphasizes the notion that synaptic transmission is both chemical and electrical, and that interactions between these two forms of interneuronal communication might be required for normal brain development and function.
Collapse
|
48
|
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: 5.5] [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
|
49
|
Kurtenbach S, Kurtenbach S, Zoidl G. Gap junction modulation and its implications for heart function. Front Physiol 2014; 5:82. [PMID: 24578694 PMCID: PMC3936571 DOI: 10.3389/fphys.2014.00082] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 02/10/2014] [Indexed: 01/04/2023] Open
Abstract
Gap junction communication (GJC) mediated by connexins is critical for heart function. To gain insight into the causal relationship of molecular mechanisms of disease pathology, it is important to understand which mechanisms contribute to impairment of gap junctional communication. Here, we present an update on the known modulators of connexins, including various interaction partners, kinases, and signaling cascades. This gap junction network (GJN) can serve as a blueprint for data mining approaches exploring the growing number of publicly available data sets from experimental and clinical studies.
Collapse
Affiliation(s)
- Stefan Kurtenbach
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| | - Sarah Kurtenbach
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| | - Georg Zoidl
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada ; Department of Biology, Faculty of Science, York University Toronto, ON, Canada ; Center for Vision Research, York University Toronto, ON, Canada
| |
Collapse
|
50
|
Nielsen MS, Axelsen LN, Sorgen PL, Verma V, Delmar M, Holstein-Rathlou NH. Gap junctions. Compr Physiol 2013; 2:1981-2035. [PMID: 23723031 DOI: 10.1002/cphy.c110051] [Citation(s) in RCA: 310] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Gap junctions are essential to the function of multicellular animals, which require a high degree of coordination between cells. In vertebrates, gap junctions comprise connexins and currently 21 connexins are known in humans. The functions of gap junctions are highly diverse and include exchange of metabolites and electrical signals between cells, as well as functions, which are apparently unrelated to intercellular communication. Given the diversity of gap junction physiology, regulation of gap junction activity is complex. The structure of the various connexins is known to some extent; and structural rearrangements and intramolecular interactions are important for regulation of channel function. Intercellular coupling is further regulated by the number and activity of channels present in gap junctional plaques. The number of connexins in cell-cell channels is regulated by controlling transcription, translation, trafficking, and degradation; and all of these processes are under strict control. Once in the membrane, channel activity is determined by the conductive properties of the connexin involved, which can be regulated by voltage and chemical gating, as well as a large number of posttranslational modifications. The aim of the present article is to review our current knowledge on the structure, regulation, function, and pharmacology of gap junctions. This will be supported by examples of how different connexins and their regulation act in concert to achieve appropriate physiological control, and how disturbances of connexin function can lead to disease.
Collapse
Affiliation(s)
- Morten Schak Nielsen
- Department of Biomedical Sciences and The Danish National Research Foundation Centre for Cardiac Arrhythmia, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|