1
|
Cibelli A, Veronica Lopez-Quintero S, Mccutcheon S, Scemes E, Spray DC, Stout RF, Suadicani SO, Thi MM, Urban-Maldonado M. Generation and Characterization of Immortalized Mouse Cortical Astrocytes From Wildtype and Connexin43 Knockout Mice. Front Cell Neurosci 2021; 15:647109. [PMID: 33790744 PMCID: PMC8005635 DOI: 10.3389/fncel.2021.647109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/18/2021] [Indexed: 11/17/2022] Open
Abstract
We transduced mouse cortical astrocytes cultured from four litters of embryonic wildtype (WT) and connexin43 (Cx43) null mouse pups with lentiviral vector encoding hTERT and measured expression of astrocyte-specific markers up to passage 10 (p10). The immortalized cell lines thus generated (designated IWCA and IKOCA, respectively) expressed biomarkers consistent with those of neonatal astrocytes, including Cx43 from wildtype but not from Cx43-null mice, lack of Cx30, and presence of Cx26. AQP4, the water channel that is found in high abundance in astrocyte end-feet, was expressed at moderately high levels in early passages, and its mRNA and protein declined to low but still detectable levels by p10. The mRNA levels of the astrocyte biomarkers aldehyde dehydrogenase 1L1 (ALDH1L1), glutamine synthetase (GS) and glial fibrillary acidic protein (GFAP) remained relatively constant during successive passages. GS protein expression was maintained while GFAP declined with cell passaging but was still detectable at p10. Both mRNA and protein levels of glutamate transporter 1 (GLT-1) declined with passage number. Immunostaining at corresponding times was consistent with the data from Western blots and provided evidence that these proteins were expressed at appropriate intracellular locations. Consistent with our goal of generating immortalized cell lines in which Cx43 was either functionally expressed or absent, IWCA cells were found to be well coupled with respect to intercellular dye transfer and similar to primary astrocyte cultures in terms of time course of junction formation, electrical coupling strength and voltage sensitivity. Moreover, barrier function was enhanced in co-culture of the IWCA cell line with bEnd.3 microvascular endothelial cells. In addition, immunostaining revealed oblate endogenous Cx43 gap junction plaques in IWCA that were similar in appearance to those plaques obtained following transfection of IKOCA cells with fluorescent protein tagged Cx43. Re-expression of Cx43 in IKOCA cells allows experimental manipulation of connexins and live imaging of interactions between connexins and other proteins. We conclude that properties of these cell lines resemble those of primary cultured astrocytes, and they may provide useful tools in functional studies by facilitating genetic and pharmacological manipulations in the context of an astrocyte-appropriate cellular environment.
Collapse
Affiliation(s)
- Antonio Cibelli
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| | | | - Sean Mccutcheon
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| | - Eliana Scemes
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| | - David C. Spray
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States,Department of Medicine (Cardiology), Albert Einstein College of Medicine, New York, NY, United States,*Correspondence: David C. Spray,
| | - Randy F. Stout
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States,Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, New York, NY, United States,Randy J. Stout Jr.,
| | - Sylvia O. Suadicani
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States,Department of Urology, Albert Einstein College of Medicine, New York, NY, United States,Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY, United States
| | - Mia M. Thi
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States,Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY, United States,Department of Orthopaedic Surgery, Albert Einstein College of Medicine, New York, NY, United States
| | - Marcia Urban-Maldonado
- Department of Urology, Albert Einstein College of Medicine, New York, NY, United States,Department of Orthopaedic Surgery, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
2
|
Tadevosyan K, Iglesias-García O, Mazo MM, Prósper F, Raya A. Engineering and Assessing Cardiac Tissue Complexity. Int J Mol Sci 2021; 22:ijms22031479. [PMID: 33540699 PMCID: PMC7867236 DOI: 10.3390/ijms22031479] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 01/28/2021] [Accepted: 01/28/2021] [Indexed: 01/14/2023] Open
Abstract
Cardiac tissue engineering is very much in a current focus of regenerative medicine research as it represents a promising strategy for cardiac disease modelling, cardiotoxicity testing and cardiovascular repair. Advances in this field over the last two decades have enabled the generation of human engineered cardiac tissue constructs with progressively increased functional capabilities. However, reproducing tissue-like properties is still a pending issue, as constructs generated to date remain immature relative to native adult heart. Moreover, there is a high degree of heterogeneity in the methodologies used to assess the functionality and cardiac maturation state of engineered cardiac tissue constructs, which further complicates the comparison of constructs generated in different ways. Here, we present an overview of the general approaches developed to generate functional cardiac tissues, discussing the different cell sources, biomaterials, and types of engineering strategies utilized to date. Moreover, we discuss the main functional assays used to evaluate the cardiac maturation state of the constructs, both at the cellular and the tissue levels. We trust that researchers interested in developing engineered cardiac tissue constructs will find the information reviewed here useful. Furthermore, we believe that providing a unified framework for comparison will further the development of human engineered cardiac tissue constructs displaying the specific properties best suited for each particular application.
Collapse
Affiliation(s)
- Karine Tadevosyan
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL) and Program for Clinical Translation of Regenerative Medicine in Catalonia (P-CMRC), 08908 L’Hospitalet del Llobregat, Spain;
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Olalla Iglesias-García
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL) and Program for Clinical Translation of Regenerative Medicine in Catalonia (P-CMRC), 08908 L’Hospitalet del Llobregat, Spain;
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, 31008 Pamplona, Spain; (M.M.M.); (F.P.)
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Correspondence: (O.I.-G.); (A.R.)
| | - Manuel M. Mazo
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, 31008 Pamplona, Spain; (M.M.M.); (F.P.)
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Hematology and Cell Therapy Area, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Felipe Prósper
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, 31008 Pamplona, Spain; (M.M.M.); (F.P.)
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
- Hematology and Cell Therapy Area, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- Center for Networked Biomedical Research on Cancer (CIBERONC), 28029 Madrid, Spain
| | - Angel Raya
- Regenerative Medicine Program, Bellvitge Institute for Biomedical Research (IDIBELL) and Program for Clinical Translation of Regenerative Medicine in Catalonia (P-CMRC), 08908 L’Hospitalet del Llobregat, Spain;
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
- Correspondence: (O.I.-G.); (A.R.)
| |
Collapse
|
3
|
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
|
4
|
Spray DC, Iglesias R, Shraer N, Suadicani SO, Belzer V, Hanstein R, Hanani M. Gap junction mediated signaling between satellite glia and neurons in trigeminal ganglia. Glia 2019; 67:791-801. [PMID: 30715764 DOI: 10.1002/glia.23554] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/28/2018] [Accepted: 10/03/2018] [Indexed: 01/07/2023]
Abstract
Peripheral sensory ganglia contain the somata of neurons mediating mechanical, thermal, and painful sensations from somatic, visceral, and oro-facial organs. Each neuronal cell body is closely surrounded by satellite glial cells (SGCs) that have properties and functions similar to those of central astrocytes, including expression of gap junction proteins and functional dye coupling. As shown in other pain models, after systemic pain induction by intra-peritoneal injection of lipopolysaccharide, dye coupling among SGCs in intact trigeminal ganglion was enhanced. Moreover, neuron-neuron and neuron-SGC coupling was also detected. To verify the presence of gap junction-mediated coupling between SGCs and sensory neurons, we performed dual whole cell patch clamp recordings from both freshly isolated and short term cultured cell pairs dissociated from mouse trigeminal ganglia. Bidirectional gap junction mediated electrical responses were frequently recorded between SGCs, between neurons and between neurons and SGCs. Polarization of SGC altered neuronal excitability, providing evidence that gap junction-mediated interactions between neurons and glia within sensory ganglia may contribute to integration of peripheral sensory responses, and to the modulation and coordinaton of neuronal activity.
Collapse
Affiliation(s)
- David C Spray
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York
| | - Rodolfo Iglesias
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York
| | - Nathanael Shraer
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York
| | - Sylvia O Suadicani
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York.,Department of Urology, Albert Einstein College of Medicine, Bronx, New York
| | - Vitali Belzer
- Laboratory of Experimental Surgery, Department of Surgery, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Regina Hanstein
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York
| | - Menachem Hanani
- Laboratory of Experimental Surgery, Department of Surgery, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
5
|
Stout RF, Spray DC. Cysteine residues in the cytoplasmic carboxy terminus of connexins dictate gap junction plaque stability. Mol Biol Cell 2017; 28:2757-2764. [PMID: 28835376 PMCID: PMC5638580 DOI: 10.1091/mbc.e17-03-0206] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 07/21/2017] [Accepted: 08/14/2017] [Indexed: 01/01/2023] Open
Abstract
Cysteine residues within the cytoplasmic carboxyl-terminus of gap junction–forming proteins are required to stabilize gap junction plaque organization. The stability of gap junction plaque organization can be modified. Gap junction stability may provide a stable supramolecular platform for modulation of gap junction functions. Gap junctions are cellular contact sites composed of clustered connexin transmembrane proteins that act in dual capacities as channels for direct intercellular exchange of small molecules and as structural adhesion complexes known as gap junction nexuses. Depending on the connexin isoform, the cluster of channels (the gap junction plaque) can be stably or fluidly arranged. Here we used confocal microscopy and mutational analysis to identify the residues within the connexin proteins that determine gap junction plaque stability. We found that stability is altered by changing redox balance using a reducing agent—indicating gap junction nexus stability is modifiable. Stability of the arrangement of connexins is thought to regulate intercellular communication by establishing an ordered supramolecular platform. By identifying the residues that establish plaque stability, these studies lay the groundwork for exploration of mechanisms by which gap junction nexus stability modulates intercellular communication.
Collapse
Affiliation(s)
- Randy F Stout
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY 11568-8000 .,Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461
| | - David C Spray
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461
| |
Collapse
|
6
|
Computational simulations of asymmetric fluxes of large molecules through gap junction channel pores. J Theor Biol 2016; 412:61-73. [PMID: 27590324 DOI: 10.1016/j.jtbi.2016.08.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 06/30/2016] [Accepted: 08/30/2016] [Indexed: 02/05/2023]
Abstract
Gap junction channels are formed out of connexin isoforms, which enable molecule and ion selective diffusion amongst neighboring cells. HeLa cells expressing distinct connexins (Cx) allow the formation of heterotypic channels, where we observed a molecular charge-independent preferential flux of large fluorescent molecules in the Cx45 to Cx43 direction. We hypothesize that the pore's shape is a significant factor along-side charge and transjunctional voltages for this asymmetric flux. To test this hypothesis, we developed a 3D computational model simulating Brownian diffusion of large molecules in a gap junction channel pore. The basic pore contour was derived from x-ray crystallographic structures of Cx43 and Cx26 and approximated using basic geometric shapes. Lucifer yellow dye molecules and cesium counter-ions were modeled as spheres using their respective Stokes radii. Our simulation results from simple diffusion and constant concentration gradient experiments showed that only charged particles yield asymmetric fluxes in heterotypic pores. While increasing the inner mouth size resulted in a near-quadratic rise in flux, the rise was asymptotic for outer mouth radii increase. Probability maps and average force per particle per pore section explain the asymmetric flux with variation in pore shape. Furthermore, the simulation results are in agreement with our in vitro experimental results with HeLa cells in Cx43-Cx45 heterotypic configurations. The presence of asymmetric fluxes can help us to understand effects of the molecular structure of the pore and predict potential differences in vivo.
Collapse
|
7
|
Coupled Activation of Primary Sensory Neurons Contributes to Chronic Pain. Neuron 2016; 91:1085-1096. [PMID: 27568517 DOI: 10.1016/j.neuron.2016.07.044] [Citation(s) in RCA: 217] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 06/21/2016] [Accepted: 07/19/2016] [Indexed: 11/20/2022]
Abstract
Primary sensory neurons in the DRG play an essential role in initiating pain by detecting painful stimuli in the periphery. Tissue injury can sensitize DRG neurons, causing heightened pain sensitivity, often leading to chronic pain. Despite the functional importance, how DRG neurons function at a population level is unclear due to the lack of suitable tools. Here we developed an imaging technique that allowed us to simultaneously monitor the activities of >1,600 neurons/DRG in live mice and discovered a striking neuronal coupling phenomenon that adjacent neurons tend to activate together following tissue injury. This coupled activation occurs among various neurons and is mediated by an injury-induced upregulation of gap junctions in glial cells surrounding DRG neurons. Blocking gap junctions attenuated neuronal coupling and mechanical hyperalgesia. Therefore, neuronal coupling represents a new form of neuronal plasticity in the DRG and contributes to pain hypersensitivity by "hijacking" neighboring neurons through gap junctions.
Collapse
|
8
|
Duchêne A, Perier M, Zhao Y, Liu X, Thomasson J, Chauveau F, Piérard C, Lagarde D, Picoli C, Jeanson T, Mouthon F, Dauvilliers Y, Giaume C, Lin JS, Charvériat M. Impact of Astroglial Connexins on Modafinil Pharmacological Properties. Sleep 2016; 39:1283-92. [PMID: 27091533 DOI: 10.5665/sleep.5854] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 02/27/2016] [Indexed: 01/11/2023] Open
Abstract
STUDY OBJECTIVES Modafinil is a non-amphetaminic wake-promoting compound used as therapy against sleepiness and narcolepsy. Its mode of action is complex, but modafinil has been recently proposed to act as a cellular-coupling enhancer in glial cells, through modulation of gap junctions constituted by connexins. The present study investigated in mice the impact of connexins on the effects of modafinil using connexin inhibitors. METHODS Modafinil was administered alone or combined with inhibitors of astrocyte connexin, meclofenamic acid, or flecainide, respectively, acting on Cx30 and Cx43. Sleep-wake states were monitored in wild-type and narcoleptic orexin knockout mice. A spontaneous alternation task was used to evaluate working memory in wild-type mice. The effects of the compounds on astroglial intercellular coupling were determined using dye transfer in acute cortical slices. RESULTS Meclofenamic acid had little modulation on the effects of modafinil, but flecainide enhanced the wake-promoting and pro-cognitive effects of modafinil. Co-administration of modafinil/flecainide resulted in a marked decrease in the number and duration of direct transitions to rapid eye movement sleep, which are characteristic of narcoleptic episodes in orexin knockout mice. Furthermore, modafinil enhanced the connexin-mediated astroglial cell coupling, whereas flecainide reduced it. Finally, this modafinil-induced effect was reversed by co-administration with flecainide. CONCLUSIONS Our study indicates that flecainide impacts the pharmacological effects of modafinil, likely through the normalization of Cx30-dependent gap junctional coupling in astroglial networks. The enhancement of the wake-promoting, behavioral, and cognitive outcomes of modafinil demonstrated here with flecainide would open new perspectives in the management of sleep disorders such as narcolepsy. COMMENTARY A commentary on this article appears in this issue on page 1175.
Collapse
Affiliation(s)
| | - Magali Perier
- Laboratory Waking, CRNL, INSERM-U1028/CNRS-UMR5292, Claude Bernard University, Lyon Cedex, France
| | - Yan Zhao
- Laboratory Waking, CRNL, INSERM-U1028/CNRS-UMR5292, Claude Bernard University, Lyon Cedex, France
| | - Xinhe Liu
- Collège de France, Centre for Interdisciplinary Research in Biology (CIRB), Paris, France
| | - Julien Thomasson
- Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Frédéric Chauveau
- Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | | | - Didier Lagarde
- Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Christèle Picoli
- Theranexus, Lyon, France.,CEA/IMETI/Theranexus, Fontenayaux-Roses, France
| | - Tiffany Jeanson
- Theranexus, Lyon, France.,Collège de France, Centre for Interdisciplinary Research in Biology (CIRB), Paris, France
| | | | - Yves Dauvilliers
- National Reference Centre for Narcolepsy, Sleep Unit, CHU Montpellier, INSERM U1061, France
| | - Christian Giaume
- Collège de France, Centre for Interdisciplinary Research in Biology (CIRB), Paris, France
| | - Jian-Sheng Lin
- Laboratory Waking, CRNL, INSERM-U1028/CNRS-UMR5292, Claude Bernard University, Lyon Cedex, France
| | | |
Collapse
|
9
|
Nguyen H, Badie N, McSpadden L, Pedrotty D, Bursac N. Quantifying electrical interactions between cardiomyocytes and other cells in micropatterned cell pairs. Methods Mol Biol 2014; 1181:249-62. [PMID: 25070342 PMCID: PMC4219610 DOI: 10.1007/978-1-4939-1047-2_21] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Micropatterning is a powerful technique to control cell shape and position on a culture substrate. In this chapter, we describe the method to reproducibly create large numbers of micropatterned heterotypic cell pairs with defined size, shape, and length of cell-cell contact. These cell pairs can be utilized in patch clamp recordings to quantify electrical interactions between cardiomyocytes and non-cardiomyocytes.
Collapse
Affiliation(s)
- Hung Nguyen
- Department of Biomedical Engineering, Duke University, 3000 Science Drive, Hudson Hall 136, 90281, Durham, NC, USA
| | | | | | | | | |
Collapse
|
10
|
Matrix-dependent adhesion mediates network responses to physiological stimulation of the osteocyte cell process. Proc Natl Acad Sci U S A 2013; 110:12096-101. [PMID: 23818616 DOI: 10.1073/pnas.1310003110] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Osteocytes are bone cells that form cellular networks that sense mechanical loads distributed throughout the bone tissue. Interstitial fluid flow in the lacunar canalicular system produces focal strains at localized attachment sites around the osteocyte cell process. These regions of periodic attachment between the osteocyte cell membrane and its canalicular wall are sites where pN-level fluid-flow induced forces are generated in vivo. In this study, we show that focally applied forces of this magnitude using a newly developed Stokesian fluid stimulus probe initiate rapid and transient intercellular electrical signals in vitro. Our experiments demonstrate both direct gap junction coupling and extracellular purinergic P2 receptor signaling between MLO-Y4 cells in a connected bone cell network. Intercellular signaling was initiated by pN-level forces applied at integrin attachment sites along both appositional and distal unapposed cell processes, but not initiated at their cell bodies with equivalent forces. Electrical coupling was evident in 58% of all cell pairs tested with appositional connections; coupling strength increased with the increasing number of junctional connections. Apyrase, a nucleotide-degrading enzyme, suppressed and abolished force-induced effector responses, indicating a contribution from ATP released by the stimulated cell. This work extends the understanding of how osteocytes modulate their microenvironment in response to mechanical signals and highlights mechanisms of intercellular relay of mechanoresponsive signals in the bone network.
Collapse
|
11
|
del Corsso C, Iglesias R, Zoidl G, Dermietzel R, Spray DC. Calmodulin dependent protein kinase increases conductance at gap junctions formed by the neuronal gap junction protein connexin36. Brain Res 2012; 1487:69-77. [PMID: 22796294 PMCID: PMC4355912 DOI: 10.1016/j.brainres.2012.06.058] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 06/26/2012] [Accepted: 06/30/2012] [Indexed: 11/27/2022]
Abstract
The major neuronal gap junction protein connexin36 (Cx36) exhibits the remarkable property of "run-up", in which junctional conductance typically increases by 10-fold or more within 5-10min following cell break-in with patch pipettes. Such conductance "run-up" is a unique property of Cx36, as it has not been seen in cell pairs expressing other connexins. Because of the recent observation describing CaMKII binding and phosphorylation sites in Cx36 and evidence that calmodulin dependent protein kinase II (CaMKII) may potentiate electrical coupling in neurons of teleosts, we have explored whether CaMKII activates mammalian Cx36. Consistent with this hypothesis, certain Cx36 mutants lacking the CaMKII binding and phosphorylation sites or wild type Cx36 treated with certain cognate peptides corresponding to binding or phosphorylation sites blocked or strongly attenuated run-up of junctional conductance. Likewise, KN-93, an inhibitor of CaMKII, blocked run-up, as did a membrane permeable peptide corresponding to the CaMKII autoinhibitory domain. Furthermore, run-up was blocked by phosphatase delivered within the pipette and not affected by treatment with the phosphatase inhibitor okadaic acid. These results imply that phosphorylation by CaMKII strengthens junctional currents of Cx36 channels, thereby conferring functional plasticity on electrical synapses formed of this protein.
Collapse
Affiliation(s)
- Cristiane del Corsso
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY,10461, USA
| | - Rodolfo Iglesias
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY,10461, USA
| | | | | | - David C. Spray
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY,10461, USA
| |
Collapse
|
12
|
Abstract
Heart attack remains the leading cause of death in both men and women worldwide. Stem cell-based therapies, including the use of engineered cardiac tissues, have the potential to treat the massive cell loss and pathological remodeling resulting from heart attack. Specifically, embryonic and induced pluripotent stem cells are a promising source for generation of therapeutically relevant numbers of functional cardiomyocytes and engineering of cardiac tissues in vitro. This review will describe methodologies for successful differentiation of pluripotent stem cells towards the cardiovascular cell lineages as they pertain to the field of cardiac tissue engineering. The emphasis will be placed on comparing the functional maturation in engineered cardiac tissues and developing heart and on methods to quantify cardiac electrical and mechanical function at different spatial scales.
Collapse
Affiliation(s)
- Brian Liau
- Department of Biomedical Engineering, Faculty of Cardiology, Duke University, Room 136 Hudson Hall, Durham, NC 27708, USA
| | | | | |
Collapse
|
13
|
Xin L, Sun Y, Bai D. Heterotypic connexin50/connexin50 mutant gap junction channels reveal interactions between two hemichannels during transjunctional voltage-dependent gating. J Physiol 2012; 590:5037-52. [PMID: 22802587 DOI: 10.1113/jphysiol.2012.235507] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
To investigate transjunctional voltage (Vj)-dependent gating mechanisms of connexin50 (Cx50) gap junction (GJ) channels and to elucidate the relative contribution of each hemichannel of a heterotypic GJ channel to Vj-dependent gating, we performed dual voltage-clamp recordings on heterotypic GJ channels formed by Cx50 and a mutant, Cx50N9R or a chimera, Cx50-Cx36N. Our results provide evidence that the two component hemichannels interact with each other during Vj-dependent gating. Cx50/Cx50N9R heterotypic GJ channels exhibited asymmetrical Vj-dependent gating which cannot be ascribed to the function of an individual hemichannel for a certain polarity of voltage; instead it can only be ascribed to the combined effects of both hemichannels. Single GJ channel open dwell-time analyses showed that homotypic Cx50 channels adopted short-lived and long-lived open states. Heterotypic combinations of Cx50/Cx50N9R gave rise to shorter mean dwell-times when Cx50-expressing cells received relatively positive Vj, and longer mean dwell-times when positive Vj was applied at the Cx50N9R side. In contrast, Cx50/Cx50-Cx36N heterotypic channels showed asymmetrical Vj-dependent gating, which appears to be caused by enhanced and reduced Vj-gating sensitivity of Cx50-Cx36N and Cx50 hemichannels, respectively. Unitary conductance of the main open state of both types of heterotypic GJ channel cannot be simply predicted by assuming a Vj redistribution across the two hemichannels arranged in series in heterotypic GJ channels. Our data also reveal reasons for the invisibility of fast Vj-gating transitions from open to substate in homotypic Cx50N9R and Cx50-Cx36N channels.
Collapse
Affiliation(s)
- Li Xin
- Graduate Program of Neuroscience, The University of Western Ontario, London, Ontario, Canada N6A 5C1
| | | | | |
Collapse
|
14
|
Picoli C, Nouvel V, Aubry F, Reboul M, Duchêne A, Jeanson T, Thomasson J, Mouthon F, Charvériat M. Human Connexin Channel Specificity of Classical and New Gap Junction Inhibitors. ACTA ACUST UNITED AC 2012; 17:1339-47. [DOI: 10.1177/1087057112452594] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Connexins are transmembrane proteins involved in gap junction intercellular communication. They present cell- and tissue-specific expression, with own electric and metabolic coupling specificities. These proteins are involved in numerous physiological processes in the brain and among them neuronal synchronization and trafficking of glucose. Such proteins are also described as being misregulated in various pathologies in the central nervous system. Thus, connexin blockers have been proposed as pharmacological tools to dissect these implications. However, such approaches lack accurate characterization of known inhibitors toward gap junction isoform specificity. In addition, those compounds are limited to few chemical classes and exhibit other activities, for example, an anti-inflammatory effect. The aims of this study were to evaluate the selectivity of described inhibitors and to enrich this pharmacopeia by new chemical classes. In this study, we present the specificity of published inhibitors toward several connexin isoforms expressed in the brain. Furthermore, after a screening of compounds using cellular models, we identified seven new inhibitors, with high functional reversibility and different relative selectivity toward isoforms. They constitute new chemical classes of connexin modulators completing those previously described. These new inhibitors might also provide new insights in understanding numerous pathophysiological processes involving gap junctions.
Collapse
Affiliation(s)
| | | | - Fabien Aubry
- Fondation Alliance BioSecure, Fondation Alliance BioSecure, Paris, France
| | - Marlène Reboul
- Fondation Alliance BioSecure, Fondation Alliance BioSecure, Paris, France
| | | | | | | | - Franck Mouthon
- CEA/IMETI/SEPIA, Fontenay-aux-Roses, France
- CEA/IMETI/THERANEXUS, Fontenay-aux-Roses, France
| | | |
Collapse
|
15
|
Iglesias RM, Spray DC. Pannexin1-mediated ATP release provides signal transmission between Neuro2A cells. Neurochem Res 2012; 37:1355-63. [PMID: 22359052 DOI: 10.1007/s11064-012-0720-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Revised: 01/13/2012] [Accepted: 01/28/2012] [Indexed: 01/22/2023]
Abstract
Pannexin1 (Panx1), a protein related to the gap junction proteins of invertebrates, forms nonjunctional channels that open upon depolarization and in response to mechanical stretch and purinergic receptor stimulation. Importantly, ATP can be released through Panx1 channels, providing a possible role for these channels in non-vesicular signal transmission. In this study we expressed exogenous human and mouse Panx1 in the gap junction deficient Neuro2A neuroblastoma cell line and explored the contribution of Panx1 channels to cell-cell communication as sites of ATP release. Electrophysiological (patch clamp) recordings from Panx1 transfected Neuro2A cells revealed membrane conductance that increased beyond 0 mV when applying voltage ramps from -60 to +100 mV; threshold was correlated with extracellular K+, so that at 10 mM K+, channels began to open at -30 mV. Evaluation of cell-cell communication using dual whole cell recordings from cell pairs revealed that activation of Panx1 current in one cell of the pair induced an inward current in the second cell after a latency of 10-20 s. This paracrine response was amplified by an ATPase inhibitor (ARL67156, 100 μM) and was blocked by the ATP-degrading enzyme apyrase (6.7 U/ml), by the P2 receptor antagonist suramin (50 μM) and by the Panx1 channel blocker carbenoxolone. These results provide additional evidence that ATP release through Panx1 channels can mediate nonsynaptic bidirectional intercellular communication. Furthermore, current potentiation by elevated K+ provides a mechanism for enhancement of ATP release under pathological conditions.
Collapse
Affiliation(s)
- Rodolfo M Iglesias
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, K 840, Bronx, NY 10461, USA
| | | |
Collapse
|
16
|
Calenda G, Suadicani SO, Iglesias R, Spray DC, Melman A, Davies KP. Silencing MaxiK activity in corporal smooth muscle cells initiates compensatory mechanisms to maintain calcium homeostasis. J Sex Med 2011; 8:2191-204. [PMID: 21269393 PMCID: PMC4337397 DOI: 10.1111/j.1743-6109.2010.02180.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION The MaxiK potassium channel is regulated by voltage and intracellular calcium, and plays a critical role in regulating intracellular calcium concentration ([Ca(2+) ](i)), which is the ultimate determinant of smooth muscle tone. Tight control of corpus cavernosum smooth muscle (CCSM) tone is critically important and misregulation can result in erectile dysfunction. AIM Because of the tight functional linkage of MaxiK and calcium channel activity, the aim of this study was to determine the effects of silencing and pharmacological inhibition of MaxiK on calcium homeostasis and intercellular calcium signaling in CCSM cells. METHODS We compared changes in the basal intracellular [Ca(2+) ](i) and parameters defining intercellular calcium wave (ICW) spread in 48 hours MaxiK silenced CCSM cells vs. acute blockade of the channel with iberiotoxin. To analyze changes occurring in gene expression we performed micro-array analysis following MaxiK silencing for 48 hours. MAIN OUTCOME MEASURES Changes in Fura-2 fluorescence intensities were measured to evaluate basal [Ca(2+) ](i) levels and ICW parameters. Microarray analysis of global gene expression was performed. RESULTS Forty-eight hours after MaxiK silencing the basal [Ca(2+) ](i) , the ICW amplitude and spread among CCSM cells were not markedly different in silenced compared to mock transfected controls, whereas short-term blockade significantly increased basal [Ca(2+) ](i) level and amplified Ca(2+) signaling among CCSM cells. Micro-array analysis showed that several genes within Ca(2+) homeostasis and smooth muscle tone regulation pathways had significantly altered expression. CONCLUSIONS Our results indicate that while short-term blockade of the MaxiK channel is associated with an increase in basal [Ca(2+) ](i), Ca(2+) homeostasis is restored during the 48 hours period following silencing. We hypothesize that the different pathways regulating [Ca(2+) ](i) and CCSM tone are linked through molecular crosstalk and that their coordinated regulation is part of a compensatory mechanism aimed to maintain Ca(2+) homeostasis and CCSM tone.
Collapse
Affiliation(s)
- Giulia Calenda
- Department of Urology, Albert Einstein College of Medicine, Bronx, New York, NY, USA
| | - Sylvia Ottilie Suadicani
- Department of Urology, Albert Einstein College of Medicine, Bronx, New York, NY, USA
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, NY, USA
| | - Rodolfo Iglesias
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, NY, USA
| | - David Conover Spray
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, NY, USA
| | - Arnold Melman
- Department of Urology, Albert Einstein College of Medicine, Bronx, New York, NY, USA
| | - Kelvin Paul Davies
- Department of Urology, Albert Einstein College of Medicine, Bronx, New York, NY, USA
| |
Collapse
|
17
|
Kirkton RD, Bursac N. Engineering biosynthetic excitable tissues from unexcitable cells for electrophysiological and cell therapy studies. Nat Commun 2011; 2:300. [PMID: 21556054 PMCID: PMC3388000 DOI: 10.1038/ncomms1302] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Accepted: 04/05/2011] [Indexed: 01/03/2023] Open
Abstract
Patch-clamp recordings in single-cell expression systems have been traditionally used to study the function of ion channels. However, this experimental setting does not enable assessment of tissue-level function such as action potential (AP) conduction. Here we introduce a biosynthetic system that permits studies of both channel activity in single cells and electrical conduction in multicellular networks. We convert unexcitable somatic cells into an autonomous source of electrically excitable and conducting cells by stably expressing only three membrane channels. The specific roles that these expressed channels have on AP shape and conduction are revealed by different pharmacological and pacing protocols. Furthermore, we demonstrate that biosynthetic excitable cells and tissues can repair large conduction defects within primary 2- and 3-dimensional cardiac cell cultures. This approach enables novel studies of ion channel function in a reproducible tissue-level setting and may stimulate the development of new cell-based therapies for excitable tissue repair.
Collapse
Affiliation(s)
- Robert D Kirkton
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | | |
Collapse
|
18
|
Bursac N, Kirkton RD, McSpadden LC, Liau B. Characterizing functional stem cell-cardiomyocyte interactions. Regen Med 2010; 5:87-105. [PMID: 20017697 DOI: 10.2217/rme.09.69] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Despite the progress in traditional pharmacological and organ transplantation therapies, heart failure still afflicts 5.3 million Americans. Since June 2000, stem cell-based approaches for the prevention and treatment of heart failure have been pursued in clinics with great excitement; however, the exact mechanisms of how transplanted cells improve heart function remain elusive. One of the main difficulties in answering these questions is the limited ability to directly access and study interactions between implanted cells and host cardiomyocytes in situ. With the growing number of candidate cell types for potential clinical use, it is becoming increasingly more important to establish standardized, well-controlled in vitro and in situ assays to compare the efficacy and safety of different stem cells in cardiac repair. This article describes recent innovative methodologies to characterize direct functional interactions between stem cells and cardiomyocytes, aimed to facilitate the rational design of future cell-based therapies for heart disease.
Collapse
Affiliation(s)
- Nenad Bursac
- Department of Biomedical Engineering, Duke University, Room 136 Hudson Hall, Durham, NC 27708, USA.
| | | | | | | |
Collapse
|
19
|
Hartveit E, Veruki ML. Accurate measurement of junctional conductance between electrically coupled cells with dual whole-cell voltage-clamp under conditions of high series resistance. J Neurosci Methods 2010; 187:13-25. [DOI: 10.1016/j.jneumeth.2009.12.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Revised: 12/03/2009] [Accepted: 12/10/2009] [Indexed: 10/20/2022]
|
20
|
Iglesias R, Spray DC, Scemes E. Mefloquine blockade of Pannexin1 currents: resolution of a conflict. CELL COMMUNICATION & ADHESION 2009; 16:131-7. [PMID: 20218915 PMCID: PMC2854254 DOI: 10.3109/15419061003642618] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The authors' laboratory has reported potent block of Pannexin1 (Panx1) currents by the antimalarial quinine derivative mefloquine. However, other laboratories have found little or no mefloquine sensitivity of Panx1 currents or processes attributable to these channels. In order to resolve this issue, the authors have performed extensive dose-response studies on Panx1-transfected neuroblastoma (Neuro2A) and rat insulinoma (Rin) cells, comparing mefloquine obtained from three suppliers and also comparing the sensitivity to diastereomers. Results indicate a 20-fold difference in sensitivity to the (-)-threo-(11R/2R) diastereomer compared to the erythro enatiomers and much lower potency of (+/-)-erythro-(R*/S*)-mefloquine obtained from one of the commercial sources. This markedly lower efficacy presumably accounts for the disparity in results from different laboratories who have applied it in Panx1 studies.
Collapse
Affiliation(s)
- Rodolfo Iglesias
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | |
Collapse
|
21
|
Patel SJ, King KR, Casali M, Yarmush ML. DNA-triggered innate immune responses are propagated by gap junction communication. Proc Natl Acad Sci U S A 2009; 106:12867-72. [PMID: 19617563 PMCID: PMC2722330 DOI: 10.1073/pnas.0809292106] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Indexed: 02/07/2023] Open
Abstract
Cells respond to infection by sensing pathogens and communicating danger signals to noninfected neighbors; however, little is known about this complex spatiotemporal process. Here we show that activation of the innate immune system by double-stranded DNA (dsDNA) triggers intercellular communication through a gap junction-dependent signaling pathway, recruiting colonies of cells to collectively secrete antiviral and inflammatory cytokines for the propagation of danger signals across the tissue at large. By using live-cell imaging of a stable IRF3-sensitive GFP reporter, we demonstrate that dsDNA sensing leads to multicellular colonies of IRF3-activated cells that express the majority of secreted cytokines, including IFNbeta and TNFalpha. Inhibiting gap junctions decreases dsDNA-induced IRF3 activation, cytokine production, and the resulting tissue-wide antiviral state, indicating that this immune response propagation pathway lies upstream of the paracrine action of secreted cytokines and may represent a host-derived mechanism for evading viral antiinterferon strategies.
Collapse
Affiliation(s)
- Suraj J. Patel
- Department of Surgery, Center for Engineering in Medicine, Massachusetts General Hospital, Boston, MA 02114
- Division of Health, Sciences, and Technology, Massachusetts Institute of Technology, Harvard Medical School, Cambridge, MA 02139; and
- Shriners Hospital for Children, Boston, MA 02114
| | - Kevin R. King
- Department of Surgery, Center for Engineering in Medicine, Massachusetts General Hospital, Boston, MA 02114
- Division of Health, Sciences, and Technology, Massachusetts Institute of Technology, Harvard Medical School, Cambridge, MA 02139; and
- Shriners Hospital for Children, Boston, MA 02114
| | - Monica Casali
- Department of Surgery, Center for Engineering in Medicine, Massachusetts General Hospital, Boston, MA 02114
- Shriners Hospital for Children, Boston, MA 02114
| | - Martin L. Yarmush
- Department of Surgery, Center for Engineering in Medicine, Massachusetts General Hospital, Boston, MA 02114
- Division of Health, Sciences, and Technology, Massachusetts Institute of Technology, Harvard Medical School, Cambridge, MA 02139; and
- Shriners Hospital for Children, Boston, MA 02114
| |
Collapse
|
22
|
Schulte JS, Scheffler A, Rojas-Gomez D, Mohr FW, Dhein S. Neonatal rat cardiomyocytes show characteristics of nonhomotypic gap junction channels. CELL COMMUNICATION & ADHESION 2008; 15:13-25. [PMID: 18649175 DOI: 10.1080/15419060802014404] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Neonatal rat cardiomyocytes mainly coexpress the connexins Cx40, Cx43, and to a small amount Cx45, leading to potential formation of mixed (heteromeric/heterotypic) gap junction channels. Using the dual-voltage clamp technique with switching clamp circuits, the authors investigated voltage sensitivity of gap junction channels between cell pairs of Cx40, Cx43, and Cx45 stably transfected HeLa cells and compared those data to data obtained from cell pairs of cultured neonatal rat cardiomyocytes. In accordance to previously published data, the relationship between normalized conductance and transjunctional voltage (g/V(j)) was quasisymmetrical for the transfected HeLa cells, indicating homotypic gap junction channels. Boltzmann curves fitted to data obtained from neonatal rat cardiomyocyte pairs expressing both Cx40 and Cx43 showed an asymmetrical inactivation pattern, which cannot be explained by the presence of pure populations of homotypic gap junction channels of either isoform. In conclusion the authors assume the additional presence of heterotypic and possibly even heteromeric gap junction channels in neonatal rat cardiomyocytes.
Collapse
Affiliation(s)
- Jan Sebastian Schulte
- Institut fur Pharmakologie und Toxikologie, Universitatsklinikum Münster, Münster, Germany.
| | | | | | | | | |
Collapse
|
23
|
Kalcheva N, Qu J, Sandeep N, Garcia L, Zhang J, Wang Z, Lampe PD, Suadicani SO, Spray DC, Fishman GI. Gap junction remodeling and cardiac arrhythmogenesis in a murine model of oculodentodigital dysplasia. Proc Natl Acad Sci U S A 2007; 104:20512-6. [PMID: 18077386 PMCID: PMC2154462 DOI: 10.1073/pnas.0705472105] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2007] [Indexed: 11/18/2022] Open
Abstract
Gap junction channels are required for normal cardiac impulse propagation, and gap junction remodeling is associated with enhanced arrhythmic risk. Oculodentodigital dysplasia (ODDD) is a multisystem syndrome due to mutations in the connexin43 (Cx43) gap junction channel gene. To determine the effects of a human connexin channelopathy on cardiac electrophysiology and arrhythmogenesis, we generated a murine model of ODDD by introducing the disease-causing I130T mutant allele into the mouse genome. Cx43 abundance was markedly reduced in mutant hearts with preferential loss of phosphorylated forms that interfered with trafficking and assembly of gap junctions in the junctional membrane. Dual whole-cell patch-clamp studies showed significantly lower junctional conductance between neonatal cell pairs from mutant hearts, and optical mapping of isolated-perfused hearts with voltage-sensitive dyes demonstrated significant slowing of conduction velocity. Programmed electrical stimulation revealed a markedly increased susceptibility to spontaneous and inducible ventricular tachyarrhythmias. In summary, our data demonstrate that the I130T mutation interferes with Cx43 posttranslational processing, resulting in diminished cell-cell coupling, slowing of impulse propagation, and a proarrhythmic substrate.
Collapse
Affiliation(s)
- Nellie Kalcheva
- *Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY 10016
| | - Jiaxiang Qu
- *Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY 10016
| | - Nefthi Sandeep
- *Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY 10016
| | - Luis Garcia
- *Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY 10016
| | - Jie Zhang
- *Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY 10016
| | - Zhiyong Wang
- *Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY 10016
| | - Paul D. Lampe
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109; and
| | - Sylvia O. Suadicani
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461
| | - David C. Spray
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Glenn I. Fishman
- *Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY 10016
| |
Collapse
|
24
|
Scemes E, Suadicani SO, Dahl G, Spray DC. Connexin and pannexin mediated cell-cell communication. NEURON GLIA BIOLOGY 2007; 3:199-208. [PMID: 18634611 PMCID: PMC2588549 DOI: 10.1017/s1740925x08000069] [Citation(s) in RCA: 179] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In this review, we briefly summarize what is known about the properties of the three families of gap junction proteins, connexins, innexins and pannexins, emphasizing their importance as intercellular channels that provide ionic and metabolic coupling and as non-junctional channels that can function as a paracrine signaling pathway. We discuss that two distinct groups of proteins form gap junctions in deuterostomes (connexins) and protostomes (innexins), and that channels formed of the deuterostome homologues of innexins (pannexins) differ from connexin channels in terms of important structural features and activation properties. These differences indicate that the two families of gap junction proteins serve distinct, complementary functions in deuterostomes. In several tissues, including the CNS, both connexins and pannexins are involved in intercellular communication, but have different roles. Connexins mainly contribute by forming the intercellular gap junction channels, which provide for junctional coupling and define the communication compartments in the CNS. We also provide new data supporting the concept that pannexins form the non-junctional channels that play paracrine roles by releasing ATP and, thus, modulating the range of the intercellular Ca(2+)-wave transmission between astrocytes in culture.
Collapse
Affiliation(s)
- Eliana Scemes
- The Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, NY, 10461, USA.
| | | | | | | |
Collapse
|