51
|
Belus MT, Rogers MA, Elzubeir A, Josey M, Rose S, Andreeva V, Yelick PC, Bates EA. Kir2.1 is important for efficient BMP signaling in mammalian face development. Dev Biol 2018; 444 Suppl 1:S297-S307. [PMID: 29571612 PMCID: PMC6148416 DOI: 10.1016/j.ydbio.2018.02.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 02/21/2018] [Accepted: 02/21/2018] [Indexed: 12/23/2022]
Abstract
Mutations that disrupt the inwardly rectifying potassium channel Kir2.1 lead to Andersen-Tawil syndrome that includes periodic paralysis, cardiac arrhythmia, cognitive deficits, craniofacial dysmorphologies and limb defects. The molecular mechanism that underlies the developmental consequences of inhibition of these channels has remained a mystery. We show that while loss of Kir2.1 function does not affect expression of several early facial patterning genes, the domain in which Pou3f3 is expressed in the maxillary arch is reduced. Pou3f3 is important for development of the jugal and squamosal bones. The reduced expression domain of Pou3f3 is consistent with the reduction in the size of the squamosal and jugal bones in Kcnj2KO/KO animals, however it does not account for the diverse craniofacial defects observed in Kcnj2KO/KO animals. We show that Kir2.1 function is required in the cranial neural crest for morphogenesis of several craniofacial structures including palate closure. We find that while the palatal shelves of Kir2.1-null embryos elevate properly, they are reduced in size due to decreased proliferation of the palatal mesenchyme. While we find no reduction in expression of BMP ligands, receptors, and associated Smads in this setting, loss of Kir2.1 reduces the efficacy of BMP signaling as shown by the reduction of phosphorylated Smad 1/5/8 and reduced expression of BMP targets Smad6 and Satb2.
Collapse
Affiliation(s)
- Matthew T Belus
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, United States
| | - Madison A Rogers
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, United States
| | - Alaaeddin Elzubeir
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, United States
| | - Megan Josey
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, United States
| | - Steven Rose
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, United States
| | - Viktoria Andreeva
- Department of Orthodontics, Division of Craniofacial and Molecular Genetics, Tufts University, Boston, MA 02111, United States
| | - Pamela C Yelick
- Department of Orthodontics, Division of Craniofacial and Molecular Genetics, Tufts University, Boston, MA 02111, United States
| | - Emily A Bates
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, United States.
| |
Collapse
|
52
|
Saadin A, Starz-Gaiano M. Cytokine exocytosis and JAK/STAT activation in the Drosophila ovary requires the vesicle trafficking regulator α-Snap. J Cell Sci 2018; 131:jcs217638. [PMID: 30404830 PMCID: PMC6288073 DOI: 10.1242/jcs.217638] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 10/11/2018] [Indexed: 01/03/2023] Open
Abstract
How vesicle trafficking components actively contribute to regulation of paracrine signaling is unclear. We genetically uncovered a requirement for α-soluble NSF attachment protein (α-Snap) in the activation of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway during Drosophila egg development. α-Snap, a well-conserved vesicle trafficking regulator, mediates association of N-ethylmaleimide-sensitive factor (NSF) and SNAREs to promote vesicle fusion. Depletion of α-Snap or the SNARE family member Syntaxin1A in epithelia blocks polar cells maintenance and prevents specification of motile border cells. Blocking apoptosis rescues polar cell maintenance in α-Snap-depleted egg chambers, indicating that the lack of border cells in mutants is due to impaired signaling. Genetic experiments implicate α-Snap and NSF in secretion of a STAT-activating cytokine. Live imaging suggests that changes in intracellular Ca2+ are linked to this event. Our data suggest a cell-type specific requirement for particular vesicle trafficking components in regulated exocytosis during development. Given the central role for STAT signaling in immunity, this work may shed light on regulation of cytokine release in humans.
Collapse
Affiliation(s)
- Afsoon Saadin
- Department of Biological Sciences, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - Michelle Starz-Gaiano
- Department of Biological Sciences, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA
| |
Collapse
|
53
|
Rusconi Trigueros R, Hopkins CR, Denton JS, Piermarini PM. Pharmacological Inhibition of Inward Rectifier Potassium Channels Induces Lethality in Larval Aedes aegypti. INSECTS 2018; 9:E163. [PMID: 30445675 PMCID: PMC6315791 DOI: 10.3390/insects9040163] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/07/2018] [Accepted: 11/12/2018] [Indexed: 11/30/2022]
Abstract
The inward rectifier potassium (Kir) channels play key roles in the physiology of mosquitoes and other insects. Our group, among others, previously demonstrated that small molecule inhibitors of Kir channels are promising lead molecules for developing new insecticides to control adult female mosquitoes. However, the potential use of Kir channel inhibitors as larvicidal agents is unknown. Here we tested the hypothesis that pharmacological inhibition of Kir channels in the larvae of Aedes aegypti, the vector of several medically important arboviruses, induces lethality. We demonstrated that adding barium, a non-specific blocker of Kir channels, or VU041, a specific small-molecule inhibitor of mosquito Kir1 channels, to the rearing water (deionized H₂O) of first instar larvae killed them within 48 h. We further showed that the toxic efficacy of VU041 within 24 h was significantly enhanced by increasing the osmolality of the rearing water to 100 mOsm/kg H₂O with NaCl, KCl or mannitol; KCl provided the strongest enhancement compared to NaCl and mannitol. These data suggest: (1) the important role of Kir channels in the acclimation of larvae to elevated ambient osmolality and KCl concentrations; and (2) the disruption of osmoregulation as a potential mechanism of the toxic action of VU041. The present study provides the first evidence that inhibition of Kir channels is lethal to larval mosquitoes and broadens the potential applications of our existing arsenal of small molecule inhibitors of Kir channels, which have previously only been considered for developing adulticides.
Collapse
Affiliation(s)
- Renata Rusconi Trigueros
- Department of Entomology, The Ohio State University, Ohio Agricultural Research and Development Center, Wooster, OH 44691, USA.
| | - Corey R Hopkins
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Jerod S Denton
- Departments of Anesthesiology and Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Peter M Piermarini
- Department of Entomology, The Ohio State University, Ohio Agricultural Research and Development Center, Wooster, OH 44691, USA.
| |
Collapse
|
54
|
Pini J, Giuliano S, Matonti J, Gannoun L, Simkin D, Rouleau M, Bendahhou S. Osteogenic and Chondrogenic Master Genes Expression Is Dependent on the Kir2.1 Potassium Channel Through the Bone Morphogenetic Protein Pathway. J Bone Miner Res 2018; 33:1826-1841. [PMID: 29813186 DOI: 10.1002/jbmr.3474] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 05/04/2018] [Accepted: 05/20/2018] [Indexed: 11/07/2022]
Abstract
Andersen's syndrome is a rare disorder affecting muscle, heart, and bone that is associated with mutations leading to a loss of function of the inwardly rectifying K+ channel Kir2.1. Although the Kir2.1 function can be anticipated in excitable cells by controlling the electrical activity, its role in non-excitable cells remains to be investigated. Using Andersen's syndrome-induced pluripotent stem cells, we investigated the cellular and molecular events during the osteoblastic and chondrogenic differentiation that are affected by the loss of the Ik1 current. We show that loss of Kir2.1 channel function impairs both osteoblastic and chondrogenic processes through the downregulation of master gene expression. This downregulation is the result of an impairment of the bone morphogenetic proteins signaling pathway through dephosphorylation of the Smad proteins. Restoring Kir2.1 channel function in Andersen's syndrome cells rescued master genes expression and restored normal osteoblast and chondrocyte behavior. Our results show that Kir2.1-mediated activity controls endochondral and intramembranous ossification signaling pathways. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Jonathan Pini
- Centre for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA, USA
| | - Serena Giuliano
- UMR7370 CNRS, LP2M, Labex ICST, University Nice Côte d'Azur, Faculté de Médecine, Nice, France
| | - Julia Matonti
- UMR7370 CNRS, LP2M, Labex ICST, University Nice Côte d'Azur, Faculté de Médecine, Nice, France
| | - Lila Gannoun
- UMR7370 CNRS, LP2M, Labex ICST, University Nice Côte d'Azur, Faculté de Médecine, Nice, France
| | - Dina Simkin
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Matthieu Rouleau
- UMR7370 CNRS, LP2M, Labex ICST, University Nice Côte d'Azur, Faculté de Médecine, Nice, France
| | - Saïd Bendahhou
- UMR7370 CNRS, LP2M, Labex ICST, University Nice Côte d'Azur, Faculté de Médecine, Nice, France
| |
Collapse
|
55
|
Piermarini PM, Inocente EA, Acosta N, Hopkins CR, Denton JS, Michel AP. Inward rectifier potassium (Kir) channels in the soybean aphid Aphis glycines: Functional characterization, pharmacology, and toxicology. JOURNAL OF INSECT PHYSIOLOGY 2018; 110:57-65. [PMID: 30196125 PMCID: PMC6173977 DOI: 10.1016/j.jinsphys.2018.09.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/05/2018] [Accepted: 09/05/2018] [Indexed: 05/21/2023]
Abstract
Inward rectifier K+ (Kir) channels contribute to a variety of physiological processes in insects and are emerging targets for insecticide development. Previous studies on insect Kir channels have primarily focused on dipteran species (e.g., mosquitoes, fruit flies). Here we identify and functionally characterize Kir channel subunits in a hemipteran insect, the soybean aphid Aphis glycines, which is an economically important insect pest and vector of soybeans. From the transcriptome and genome of Ap. glycines we identified two cDNAs, ApKir1 and ApKir2, encoding Kir subunits that were orthologs of insect Kir1 and Kir2, respectively. Notably, a gene encoding a Kir3 subunit was absent from the transcriptome and genome of Ap. glycines, similar to the pea aphid Acyrthosiphon pisum. Heterologous expression of ApKir1 and ApKir2 in Xenopus laevis oocytes enhanced K+-currents in the plasma membrane; these currents were inhibited by barium and the small molecule VU041. Compared to ApKir2, ApKir1 mediated currents that were larger in magnitude, more sensitive to barium, and less inhibited by small molecule VU041. Moreover, ApKir1 exhibited stronger inward rectification compared to ApKir2. Topical application of VU041 in adult aphids resulted in dose-dependent mortality within 24 h that was more efficacious than flonicamid, an established insecticide also known to inhibit Kir channels. We conclude that despite the apparent loss of Kir3 genes in aphid evolution, Kir channels are important to aphid survival and represent a promising target for the development of new insecticides.
Collapse
Affiliation(s)
- Peter M Piermarini
- Department of Entomology, The Ohio State University, Ohio Agricultural Research and Development Center, Wooster, OH, USA.
| | - Edna Alfaro Inocente
- Department of Entomology, The Ohio State University, Ohio Agricultural Research and Development Center, Wooster, OH, USA
| | - Nuris Acosta
- Department of Entomology, The Ohio State University, Ohio Agricultural Research and Development Center, Wooster, OH, USA
| | - Corey R Hopkins
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jerod S Denton
- Departments of Anesthesiology and Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Andrew P Michel
- Department of Entomology, The Ohio State University, Ohio Agricultural Research and Development Center, Wooster, OH, USA
| |
Collapse
|
56
|
Bioelectrical coupling in multicellular domains regulated by gap junctions: A conceptual approach. Bioelectrochemistry 2018; 123:45-61. [DOI: 10.1016/j.bioelechem.2018.04.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/13/2018] [Accepted: 04/17/2018] [Indexed: 12/16/2022]
|
57
|
Laugel-Haushalter V, Morkmued S, Stoetzel C, Geoffroy V, Muller J, Boland A, Deleuze JF, Chennen K, Pitiphat W, Dollfus H, Niederreither K, Bloch-Zupan A, Pungchanchaikul P. Genetic Evidence Supporting the Role of the Calcium Channel, CACNA1S, in Tooth Cusp and Root Patterning. Front Physiol 2018; 9:1329. [PMID: 30319441 PMCID: PMC6170876 DOI: 10.3389/fphys.2018.01329] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 09/03/2018] [Indexed: 01/08/2023] Open
Abstract
In this study, we report a unique dominantly inherited disorganized supernumerary cusp and single root phenotype presented by 11 affected individuals belonging to 5 north-eastern Thai families. Using whole exome sequencing (WES) we identified a common single missense mutation that segregates with the phenotype in exon 6 of CACNA1S (Cav1.1) (NM_000069.2: c.[865A > G];[=] p.[Ile289Val];[=]), the Calcium Channel, Voltage-Dependent, L Type, Alpha-1s Subunit, OMIM ∗ 114208), affecting a highly conserved amino-acid isoleucine residue within the pore forming subdomain of CACNA1S protein. This is a strong genetic evidence that a voltage-dependent calcium ion channel is likely to play a role in influencing tooth morphogenesis and patterning.
Collapse
Affiliation(s)
- Virginie Laugel-Haushalter
- Laboratoire de Génétique Médicale, UMR_S INSERM U1112, IGMA, Faculté de Médecine, FMTS, Université de Strasbourg, Strasbourg, France
| | - Supawich Morkmued
- Biofilm Research Group, Department of Pediatric Dentistry, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand.,Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CERBM, INSERM U 1258, CNRS- UMR 7104, Université de Strasbourg, Strasbourg, France
| | - Corinne Stoetzel
- Laboratoire de Génétique Médicale, UMR_S INSERM U1112, IGMA, Faculté de Médecine, FMTS, Université de Strasbourg, Strasbourg, France
| | - Véronique Geoffroy
- Laboratoire de Génétique Médicale, UMR_S INSERM U1112, IGMA, Faculté de Médecine, FMTS, Université de Strasbourg, Strasbourg, France
| | - Jean Muller
- Laboratoire de Génétique Médicale, UMR_S INSERM U1112, IGMA, Faculté de Médecine, FMTS, Université de Strasbourg, Strasbourg, France.,Laboratoires de Diagnostic Génétique, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Anne Boland
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, Direction de la Recherche Fondamentale, Commissariat à l'Energie Atomique et aux Energies Alternatives, Paris, France
| | - Jean-François Deleuze
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, Direction de la Recherche Fondamentale, Commissariat à l'Energie Atomique et aux Energies Alternatives, Paris, France
| | - Kirsley Chennen
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CERBM, INSERM U 1258, CNRS- UMR 7104, Université de Strasbourg, Strasbourg, France.,Department of Computer Science, ICube, CNRS - UMR 7357, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Waranuch Pitiphat
- Department of Community Dentistry, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand
| | - Hélène Dollfus
- Laboratoire de Génétique Médicale, UMR_S INSERM U1112, IGMA, Faculté de Médecine, FMTS, Université de Strasbourg, Strasbourg, France.,Centre de Référence pour les Affections Rares en Génétique Ophtalmologique, Filière SENSGENE, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Karen Niederreither
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CERBM, INSERM U 1258, CNRS- UMR 7104, Université de Strasbourg, Strasbourg, France.,Faculté de Chirurgie Dentaire, Université de Strasbourg, Strasbourg, France
| | - Agnès Bloch-Zupan
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CERBM, INSERM U 1258, CNRS- UMR 7104, Université de Strasbourg, Strasbourg, France.,Faculté de Chirurgie Dentaire, Université de Strasbourg, Strasbourg, France.,Hôpitaux Universitaires de Strasbourg (HUS), Pôle de Médecine et Chirurgie Bucco-Dentaires Hôpital Civil, Centre de Référence des Maladies Rares Orales et Dentaires, O-Rares, Filière Santé Maladies Rares TETE COU, European Reference Network ERN CRANIO, Strasbourg, France
| | - Patimaporn Pungchanchaikul
- Biofilm Research Group, Department of Pediatric Dentistry, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
58
|
Yin W, Kim HT, Wang S, Gunawan F, Wang L, Kishimoto K, Zhong H, Roman D, Preussner J, Guenther S, Graef V, Buettner C, Grohmann B, Looso M, Morimoto M, Mardon G, Offermanns S, Stainier DYR. The potassium channel KCNJ13 is essential for smooth muscle cytoskeletal organization during mouse tracheal tubulogenesis. Nat Commun 2018; 9:2815. [PMID: 30022023 PMCID: PMC6052067 DOI: 10.1038/s41467-018-05043-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 05/25/2018] [Indexed: 12/22/2022] Open
Abstract
Tubulogenesis is essential for the formation and function of internal organs. One such organ is the trachea, which allows gas exchange between the external environment and the lungs. However, the cellular and molecular mechanisms underlying tracheal tube development remain poorly understood. Here, we show that the potassium channel KCNJ13 is a critical modulator of tracheal tubulogenesis. We identify Kcnj13 in an ethylnitrosourea forward genetic screen for regulators of mouse respiratory organ development. Kcnj13 mutants exhibit a shorter trachea as well as defective smooth muscle (SM) cell alignment and polarity. KCNJ13 is essential to maintain ion homeostasis in tracheal SM cells, which is required for actin polymerization. This process appears to be mediated, at least in part, through activation of the actin regulator AKT, as pharmacological increase of AKT phosphorylation ameliorates the Kcnj13-mutant trachea phenotypes. These results provide insight into the role of ion homeostasis in cytoskeletal organization during tubulogenesis.
Collapse
Affiliation(s)
- Wenguang Yin
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany.
| | - Hyun-Taek Kim
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - ShengPeng Wang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Felix Gunawan
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Lei Wang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Keishi Kishimoto
- Laboratory for Lung Development, RIKEN Center for Developmental Biology, Kobe, 650-0047, Japan
| | - Hua Zhong
- Departments of Pathology and Immunology and Molecular and Human Genetics, Integrative Molecular and Biomedical Sciences Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Dany Roman
- Departments of Pathology and Immunology and Molecular and Human Genetics, Integrative Molecular and Biomedical Sciences Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jens Preussner
- Max Planck Institute for Heart and Lung Research, ECCPS Bioinformatics and Deep Sequencing Platform, Bad Nauheim, 61231, Germany
| | - Stefan Guenther
- Max Planck Institute for Heart and Lung Research, ECCPS Bioinformatics and Deep Sequencing Platform, Bad Nauheim, 61231, Germany
| | - Viola Graef
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Carmen Buettner
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Beate Grohmann
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Mario Looso
- Max Planck Institute for Heart and Lung Research, ECCPS Bioinformatics and Deep Sequencing Platform, Bad Nauheim, 61231, Germany
| | - Mitsuru Morimoto
- Laboratory for Lung Development, RIKEN Center for Developmental Biology, Kobe, 650-0047, Japan
| | - Graeme Mardon
- Departments of Pathology and Immunology and Molecular and Human Genetics, Integrative Molecular and Biomedical Sciences Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
- Center for Molecular Medicine, Goethe University, Frankfurt, 60590, Germany
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany.
| |
Collapse
|
59
|
Bioelectric-calcineurin signaling module regulates allometric growth and size of the zebrafish fin. Sci Rep 2018; 8:10391. [PMID: 29991812 PMCID: PMC6039437 DOI: 10.1038/s41598-018-28450-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/18/2018] [Indexed: 01/01/2023] Open
Abstract
The establishment of relative size of organs and structures is paramount for attaining final form and function of an organism. Importantly, variation in the proportions of structures frequently underlies adaptive change in morphology in evolution and maybe a common mechanism underlying selection. However, the mechanism by which growth is integrated within tissues during development to achieve proper proportionality is poorly understood. We have shown that signaling by potassium channels mediates coordinated size regulation in zebrafish fins. Recently, calcineurin inhibitors were shown to elicit changes in zebrafish fin allometry as well. Here, we identify the potassium channel kcnk5b as a key player in integrating calcineurin’s growth effects, in part through regulation of the cytoplasmic C-terminus of the channel. We propose that the interaction between Kcnk5b and calcineurin acts as a signaling node to regulate allometric growth. Importantly, we find that this regulation is epistatic to inherent mechanisms instructing overall size as inhibition of calcineurin is able to bypass genetic instruction of size as seen in sof and wild-type fins, however, it is not sufficient to re-specify positional memory of size of the fin. These findings integrate classic signaling mediators such as calcineurin with ion channel function in the regulation of size and proportion during growth.
Collapse
|
60
|
Levin M, Martyniuk CJ. The bioelectric code: An ancient computational medium for dynamic control of growth and form. Biosystems 2018; 164:76-93. [PMID: 28855098 PMCID: PMC10464596 DOI: 10.1016/j.biosystems.2017.08.009] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 08/20/2017] [Accepted: 08/22/2017] [Indexed: 12/19/2022]
Abstract
What determines large-scale anatomy? DNA does not directly specify geometrical arrangements of tissues and organs, and a process of encoding and decoding for morphogenesis is required. Moreover, many species can regenerate and remodel their structure despite drastic injury. The ability to obtain the correct target morphology from a diversity of initial conditions reveals that the morphogenetic code implements a rich system of pattern-homeostatic processes. Here, we describe an important mechanism by which cellular networks implement pattern regulation and plasticity: bioelectricity. All cells, not only nerves and muscles, produce and sense electrical signals; in vivo, these processes form bioelectric circuits that harness individual cell behaviors toward specific anatomical endpoints. We review emerging progress in reading and re-writing anatomical information encoded in bioelectrical states, and discuss the approaches to this problem from the perspectives of information theory, dynamical systems, and computational neuroscience. Cracking the bioelectric code will enable much-improved control over biological patterning, advancing basic evolutionary developmental biology as well as enabling numerous applications in regenerative medicine and synthetic bioengineering.
Collapse
Affiliation(s)
- Michael Levin
- Allen Discovery Center at Tufts University, Biology Department, Tufts University, 200 Boston Avenue, Suite 4600 Medford, MA 02155, USA.
| | - Christopher J Martyniuk
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32611, USA
| |
Collapse
|
61
|
Chen R, Swale DR. Inwardly Rectifying Potassium (Kir) Channels Represent a Critical Ion Conductance Pathway in the Nervous Systems of Insects. Sci Rep 2018; 8:1617. [PMID: 29371678 PMCID: PMC5785497 DOI: 10.1038/s41598-018-20005-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/10/2018] [Indexed: 12/13/2022] Open
Abstract
A complete understanding of the physiological pathways critical for proper function of the insect nervous system is still lacking. The recent development of potent and selective small-molecule modulators of insect inward rectifier potassium (Kir) channels has enabled the interrogation of the physiological role and toxicological potential of Kir channels within various insect tissue systems. Therefore, we aimed to highlight the physiological and functional role of neural Kir channels the central nervous system, muscular system, and neuromuscular system through pharmacological and genetic manipulations. Our data provide significant evidence that Drosophila neural systems rely on the inward conductance of K+ ions for proper function since pharmacological inhibition and genetic ablation of neural Kir channels yielded dramatic alterations of the CNS spike discharge frequency and broadening and reduced amplitude of the evoked EPSP at the neuromuscular junction. Based on these data, we conclude that neural Kir channels in insects (1) are critical for proper function of the insect nervous system, (2) represents an unexplored physiological pathway that is likely to shape the understanding of neuronal signaling, maintenance of membrane potentials, and maintenance of the ionic balance of insects, and (3) are capable of inducing acute toxicity to insects through neurological poisoning.
Collapse
Affiliation(s)
- Rui Chen
- Louisiana State University AgCenter, Department of Entomology, Baton Rouge, LA, 70803, USA
| | - Daniel R Swale
- Louisiana State University AgCenter, Department of Entomology, Baton Rouge, LA, 70803, USA.
| |
Collapse
|
62
|
McLaughlin KA, Levin M. Bioelectric signaling in regeneration: Mechanisms of ionic controls of growth and form. Dev Biol 2018; 433:177-189. [PMID: 29291972 PMCID: PMC5753428 DOI: 10.1016/j.ydbio.2017.08.032] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/23/2017] [Accepted: 08/28/2017] [Indexed: 12/11/2022]
Abstract
The ability to control pattern formation is critical for the both the embryonic development of complex structures as well as for the regeneration/repair of damaged or missing tissues and organs. In addition to chemical gradients and gene regulatory networks, endogenous ion flows are key regulators of cell behavior. Not only do bioelectric cues provide information needed for the initial development of structures, they also enable the robust restoration of normal pattern after injury. In order to expand our basic understanding of morphogenetic processes responsible for the repair of complex anatomy, we need to identify the roles of endogenous voltage gradients, ion flows, and electric fields. In complement to the current focus on molecular genetics, decoding the information transduced by bioelectric cues enhances our knowledge of the dynamic control of growth and pattern formation. Recent advances in science and technology place us in an exciting time to elucidate the interplay between molecular-genetic inputs and important biophysical cues that direct the creation of tissues and organs. Moving forward, these new insights enable additional approaches to direct cell behavior and may result in profound advances in augmentation of regenerative capacity.
Collapse
Affiliation(s)
- Kelly A McLaughlin
- Allen Discovery Center, Department of Biology, Tufts University, 200 Boston Ave., Suite 4700, Medford, MA 02155, United States.
| | - Michael Levin
- Allen Discovery Center, Department of Biology, Tufts University, 200 Boston Ave., Suite 4700, Medford, MA 02155, United States
| |
Collapse
|
63
|
Kennedy T, Broadie K. Fragile X Mental Retardation Protein Restricts Small Dye Iontophoresis Entry into Central Neurons. J Neurosci 2017; 37:9844-9858. [PMID: 28887386 PMCID: PMC5637114 DOI: 10.1523/jneurosci.0723-17.2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 07/27/2017] [Accepted: 08/29/2017] [Indexed: 01/29/2023] Open
Abstract
Fragile X mental retardation protein (FMRP) loss causes Fragile X syndrome (FXS), a major disorder characterized by autism, intellectual disability, hyperactivity, and seizures. FMRP is both an RNA- and channel-binding regulator, with critical roles in neural circuit formation and function. However, it remains unclear how these FMRP activities relate to each other and how dysfunction in their absence underlies FXS neurological symptoms. In testing circuit level defects in the Drosophila FXS model, we discovered a completely unexpected and highly robust neuronal dye iontophoresis phenotype in the well mapped giant fiber (GF) circuit. Controlled dye injection into the GF interneuron results in a dramatic increase in dye uptake in neurons lacking FMRP. Transgenic wild-type FMRP reintroduction rescues the mutant defect, demonstrating a specific FMRP requirement. This phenotype affects only small dyes, but is independent of dye charge polarity. Surprisingly, the elevated dye iontophoresis persists in shaking B mutants that eliminate gap junctions and dye coupling among GF circuit neurons. We therefore used a wide range of manipulations to investigate the dye uptake defect, including timed injection series, pharmacology and ion replacement, and optogenetic activity studies. The results show that FMRP strongly limits the rate of dye entry via a cytosolic mechanism. This study reveals an unexpected new phenotype in a physical property of central neurons lacking FMRP that could underlie aspects of FXS disruption of neural function.SIGNIFICANCE STATEMENT FXS is a leading heritable cause of intellectual disability and autism spectrum disorders. Although researchers established the causal link with FMRP loss >;25 years ago, studies continue to reveal diverse FMRP functions. The Drosophila FXS model is key to discovering new FMRP roles, because of its genetic malleability and individually identified neuron maps. Taking advantage of a well characterized Drosophila neural circuit, we discovered that neurons lacking FMRP take up dramatically more current-injected small dye. After examining many neuronal properties, we determined that this dye defect is cytoplasmic and occurs due to a highly elevated dye iontophoresis rate. We also report several new factors affecting neuron dye uptake. Understanding how FMRP regulates iontophoresis should reveal new molecular factors underpinning FXS dysfunction.
Collapse
Affiliation(s)
| | - Kendal Broadie
- Department of Biological Sciences,
- Department of Cell and Developmental Biology, and
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, Tennessee 37235
| |
Collapse
|
64
|
Moore D, Walker SI, Levin M. Cancer as a disorder of patterning information: computational and biophysical perspectives on the cancer problem. CONVERGENT SCIENCE PHYSICAL ONCOLOGY 2017. [DOI: 10.1088/2057-1739/aa8548] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
65
|
Regulation of Drosophila hematopoietic sites by Activin-β from active sensory neurons. Nat Commun 2017; 8:15990. [PMID: 28748922 PMCID: PMC5537569 DOI: 10.1038/ncomms15990] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 05/23/2017] [Indexed: 12/21/2022] Open
Abstract
An outstanding question in animal development, tissue homeostasis and disease is how cell populations adapt to sensory inputs. During Drosophila larval development, hematopoietic sites are in direct contact with sensory neuron clusters of the peripheral nervous system (PNS), and blood cells (hemocytes) require the PNS for their survival and recruitment to these microenvironments, known as Hematopoietic Pockets. Here we report that Activin-β, a TGF-β family ligand, is expressed by sensory neurons of the PNS and regulates the proliferation and adhesion of hemocytes. These hemocyte responses depend on PNS activity, as shown by agonist treatment and transient silencing of sensory neurons. Activin-β has a key role in this regulation, which is apparent from reporter expression and mutant analyses. This mechanism of local sensory neurons controlling blood cell adaptation invites evolutionary parallels with vertebrate hematopoietic progenitors and the independent myeloid system of tissue macrophages, whose regulation by local microenvironments remain undefined.
Collapse
|
66
|
Dahal GR, Pradhan SJ, Bates EA. Inwardly rectifying potassium channels influence Drosophila wing morphogenesis by regulating Dpp release. Development 2017; 144:2771-2783. [PMID: 28684627 DOI: 10.1242/dev.146647] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 06/14/2017] [Indexed: 12/23/2022]
Abstract
Loss of embryonic ion channel function leads to morphological defects, but the underlying reason for these defects remains elusive. Here, we show that inwardly rectifying potassium (Irk) channels regulate release of the Drosophila bone morphogenetic protein Dpp in the developing fly wing and that this is necessary for developmental signaling. Inhibition of Irk channels decreases the incidence of distinct Dpp-GFP release events above baseline fluorescence while leading to a broader distribution of Dpp-GFP. Work by others in different cell types has shown that Irk channels regulate peptide release by modulating membrane potential and calcium levels. We found calcium transients in the developing wing, and inhibition of Irk channels reduces the duration and amplitude of calcium transients. Depolarization with high extracellular potassium evokes Dpp release. Taken together, our data implicate Irk channels as a requirement for regulated release of Dpp, highlighting the importance of the temporal pattern of Dpp presentation for morphogenesis of the wing.
Collapse
Affiliation(s)
- Giri Raj Dahal
- University of Colorado Denver School of Medicine, 12800 E 19th Avenue, Aurora, CO 80045, USA
| | - Sarala Joshi Pradhan
- University of Colorado Denver School of Medicine, 12800 E 19th Avenue, Aurora, CO 80045, USA
| | - Emily Anne Bates
- University of Colorado Denver School of Medicine, 12800 E 19th Avenue, Aurora, CO 80045, USA
| |
Collapse
|
67
|
Levin M, Pezzulo G, Finkelstein JM. Endogenous Bioelectric Signaling Networks: Exploiting Voltage Gradients for Control of Growth and Form. Annu Rev Biomed Eng 2017; 19:353-387. [PMID: 28633567 PMCID: PMC10478168 DOI: 10.1146/annurev-bioeng-071114-040647] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Living systems exhibit remarkable abilities to self-assemble, regenerate, and remodel complex shapes. How cellular networks construct and repair specific anatomical outcomes is an open question at the heart of the next-generation science of bioengineering. Developmental bioelectricity is an exciting emerging discipline that exploits endogenous bioelectric signaling among many cell types to regulate pattern formation. We provide a brief overview of this field, review recent data in which bioelectricity is used to control patterning in a range of model systems, and describe the molecular tools being used to probe the role of bioelectrics in the dynamic control of complex anatomy. We suggest that quantitative strategies recently developed to infer semantic content and information processing from ionic activity in the brain might provide important clues to cracking the bioelectric code. Gaining control of the mechanisms by which large-scale shape is regulated in vivo will drive transformative advances in bioengineering, regenerative medicine, and synthetic morphology, and could be used to therapeutically address birth defects, traumatic injury, and cancer.
Collapse
Affiliation(s)
- Michael Levin
- Biology Department, Tufts University, Medford, Massachusetts 02155-4243;
- Allen Discovery Center, Tufts University, Medford, Massachusetts 02155;
| | - Giovanni Pezzulo
- Institute of Cognitive Sciences and Technologies, National Research Council, Rome 00185, Italy;
| | | |
Collapse
|
68
|
Hooper JE, Feng W, Li H, Leach SM, Phang T, Siska C, Jones KL, Spritz RA, Hunter LE, Williams T. Systems biology of facial development: contributions of ectoderm and mesenchyme. Dev Biol 2017; 426:97-114. [PMID: 28363736 PMCID: PMC5530582 DOI: 10.1016/j.ydbio.2017.03.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 03/23/2017] [Accepted: 03/23/2017] [Indexed: 12/17/2022]
Abstract
The rapid increase in gene-centric biological knowledge coupled with analytic approaches for genomewide data integration provides an opportunity to develop systems-level understanding of facial development. Experimental analyses have demonstrated the importance of signaling between the surface ectoderm and the underlying mesenchyme are coordinating facial patterning. However, current transcriptome data from the developing vertebrate face is dominated by the mesenchymal component, and the contributions of the ectoderm are not easily identified. We have generated transcriptome datasets from critical periods of mouse face formation that enable gene expression to be analyzed with respect to time, prominence, and tissue layer. Notably, by separating the ectoderm and mesenchyme we considerably improved the sensitivity compared to data obtained from whole prominences, with more genes detected over a wider dynamic range. From these data we generated a detailed description of ectoderm-specific developmental programs, including pan-ectodermal programs, prominence- specific programs and their temporal dynamics. The genes and pathways represented in these programs provide mechanistic insights into several aspects of ectodermal development. We also used these data to identify co-expression modules specific to facial development. We then used 14 co-expression modules enriched for genes involved in orofacial clefts to make specific mechanistic predictions about genes involved in tongue specification, in nasal process patterning and in jaw development. Our multidimensional gene expression dataset is a unique resource for systems analysis of the developing face; our co-expression modules are a resource for predicting functions of poorly annotated genes, or for predicting roles for genes that have yet to be studied in the context of facial development; and our analytic approaches provide a paradigm for analysis of other complex developmental programs.
Collapse
Affiliation(s)
- Joan E Hooper
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, 12801 E 17th Avenue, Aurora, CO 80045, USA; Computational Bioscience Program, University of Colorado School of Medicine, 12801 E 17th Avenue, Aurora, CO 80045, USA.
| | - Weiguo Feng
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, 12801 E 17th Avenue, Aurora, CO 80045, USA; Department of Craniofacial Biology, University of Colorado School of Dental Medicine, 12801 E 17th Avenue, Aurora, CO 80045, USA.
| | - Hong Li
- Department of Craniofacial Biology, University of Colorado School of Dental Medicine, 12801 E 17th Avenue, Aurora, CO 80045, USA.
| | - Sonia M Leach
- Department of Biomedical Research, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA.
| | - Tzulip Phang
- Computational Bioscience Program, University of Colorado School of Medicine, 12801 E 17th Avenue, Aurora, CO 80045, USA; Department of Medicine, University of Colorado School of Medicine, 12801 E 17th Avenue, Aurora, CO 80045, USA.
| | - Charlotte Siska
- Computational Bioscience Program, University of Colorado School of Medicine, 12801 E 17th Avenue, Aurora, CO 80045, USA.
| | - Kenneth L Jones
- Department of Pediatrics, University of Colorado School of Medicine, 12801 E 17th Avenue, Aurora, CO 80045, USA.
| | - Richard A Spritz
- Human Medical Genetics and Genomics Program, University of Colorado School of Medicine, 12800 E 17th Avenue, Aurora, CO 80045, USA.
| | - Lawrence E Hunter
- Computational Bioscience Program, University of Colorado School of Medicine, 12801 E 17th Avenue, Aurora, CO 80045, USA; Department of Pharmacology, University of Colorado School of Medicine, 12801 E 17th Avenue, Aurora, CO 80045, USA.
| | - Trevor Williams
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, 12801 E 17th Avenue, Aurora, CO 80045, USA; Department of Craniofacial Biology, University of Colorado School of Dental Medicine, 12801 E 17th Avenue, Aurora, CO 80045, USA.
| |
Collapse
|
69
|
Durant F, Morokuma J, Fields C, Williams K, Adams DS, Levin M. Long-Term, Stochastic Editing of Regenerative Anatomy via Targeting Endogenous Bioelectric Gradients. Biophys J 2017; 112:2231-2243. [PMID: 28538159 PMCID: PMC5443973 DOI: 10.1016/j.bpj.2017.04.011] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/30/2017] [Accepted: 04/14/2017] [Indexed: 12/22/2022] Open
Abstract
We show that regenerating planarians' normal anterior-posterior pattern can be permanently rewritten by a brief perturbation of endogenous bioelectrical networks. Temporary modulation of regenerative bioelectric dynamics in amputated trunk fragments of planaria stochastically results in a constant ratio of regenerates with two heads to regenerates with normal morphology. Remarkably, this is shown to be due not to partial penetrance of treatment, but a profound yet hidden alteration to the animals' patterning circuitry. Subsequent amputations of the morphologically normal regenerates in water result in the same ratio of double-headed to normal morphology, revealing a cryptic phenotype that is not apparent unless the animals are cut. These animals do not differ from wild-type worms in histology, expression of key polarity genes, or neoblast distribution. Instead, the altered regenerative bodyplan is stored in seemingly normal planaria via global patterns of cellular resting potential. This gradient is functionally instructive, and represents a multistable, epigenetic anatomical switch: experimental reversals of bioelectric state reset subsequent regenerative morphology back to wild-type. Hence, bioelectric properties can stably override genome-default target morphology, and provide a tractable control point for investigating cryptic phenotypes and the stochasticity of large-scale epigenetic controls.
Collapse
Affiliation(s)
- Fallon Durant
- Allen Discovery Center at Tufts University, and Department of Biology, Tufts University, Medford, Massachusetts
| | - Junji Morokuma
- Allen Discovery Center at Tufts University, and Department of Biology, Tufts University, Medford, Massachusetts
| | | | - Katherine Williams
- Allen Discovery Center at Tufts University, and Department of Biology, Tufts University, Medford, Massachusetts
| | - Dany Spencer Adams
- Allen Discovery Center at Tufts University, and Department of Biology, Tufts University, Medford, Massachusetts
| | - Michael Levin
- Allen Discovery Center at Tufts University, and Department of Biology, Tufts University, Medford, Massachusetts.
| |
Collapse
|
70
|
Pitcairn E, Harris H, Epiney J, Pai VP, Lemire JM, Ye B, Shi NQ, Levin M, McLaughlin KA. Coordinating heart morphogenesis: A novel role for hyperpolarization-activated cyclic nucleotide-gated (HCN) channels during cardiogenesis in Xenopus laevis. Commun Integr Biol 2017; 10:e1309488. [PMID: 28702127 PMCID: PMC5501196 DOI: 10.1080/19420889.2017.1309488] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 03/16/2017] [Indexed: 12/18/2022] Open
Abstract
Hyperpolarization-activated cyclic-nucleotide gated channel (HCN) proteins are important regulators of both neuronal and cardiac excitability. Among the 4 HCN isoforms, HCN4 is known as a pacemaker channel, because it helps control the periodicity of contractions in vertebrate hearts. Although the physiological role of HCN4 channel has been studied in adult mammalian hearts, an earlier role during embryogenesis has not been clearly established. Here, we probe the embryonic roles of HCN4 channels, providing the first characterization of the expression profile of any of the HCN isoforms during Xenopus laevis development and investigate the consequences of altering HCN4 function on embryonic pattern formation. We demonstrate that both overexpression of HCN4 and injection of dominant-negative HCN4 mRNA during early embryogenesis results in improper expression of key patterning genes and severely malformed hearts. Our results suggest that HCN4 serves to coordinate morphogenetic control factors that provide positional information during heart morphogenesis in Xenopus.
Collapse
Affiliation(s)
- Emily Pitcairn
- Department of Biology and Allen Discovery Center at Tufts University, Medford, MA, USA
| | - Hannah Harris
- Department of Biology and Allen Discovery Center at Tufts University, Medford, MA, USA
| | - Justine Epiney
- Department of Biology and Allen Discovery Center at Tufts University, Medford, MA, USA
| | - Vaibhav P Pai
- Department of Biology and Allen Discovery Center at Tufts University, Medford, MA, USA
| | - Joan M Lemire
- Department of Biology and Allen Discovery Center at Tufts University, Medford, MA, USA
| | - Bin Ye
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Nian-Qing Shi
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael Levin
- Department of Biology and Allen Discovery Center at Tufts University, Medford, MA, USA
| | - Kelly A McLaughlin
- Department of Biology and Allen Discovery Center at Tufts University, Medford, MA, USA
| |
Collapse
|
71
|
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: 63] [Impact Index Per Article: 7.9] [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
|
72
|
Swale DR, Engers DW, Bollinger SR, Gross A, Inocente EA, Days E, Kanga F, Johnson RM, Yang L, Bloomquist JR, Hopkins CR, Piermarini PM, Denton JS. An insecticide resistance-breaking mosquitocide targeting inward rectifier potassium channels in vectors of Zika virus and malaria. Sci Rep 2016; 6:36954. [PMID: 27849039 PMCID: PMC5111108 DOI: 10.1038/srep36954] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 10/19/2016] [Indexed: 01/01/2023] Open
Abstract
Insecticide resistance is a growing threat to mosquito control programs around the world, thus creating the need to discover novel target sites and target-specific compounds for insecticide development. Emerging evidence suggests that mosquito inward rectifier potassium (Kir) channels represent viable molecular targets for developing insecticides with new mechanisms of action. Here we describe the discovery and characterization of VU041, a submicromolar-affinity inhibitor of Anopheles (An.) gambiae and Aedes (Ae.) aegypti Kir1 channels that incapacitates adult female mosquitoes from representative insecticide-susceptible and -resistant strains of An. gambiae (G3 and Akron, respectively) and Ae. aegypti (Liverpool and Puerto Rico, respectively) following topical application. VU041 is selective for mosquito Kir channels over several mammalian orthologs, with the exception of Kir2.1, and is not lethal to honey bees. Medicinal chemistry was used to develop an analog, termed VU730, which retains activity toward mosquito Kir1 but is not active against Kir2.1 or other mammalian Kir channels. Thus, VU041 and VU730 are promising chemical scaffolds for developing new classes of insecticides to combat insecticide-resistant mosquitoes and the transmission of mosquito-borne diseases, such as Zika virus, without harmful effects on humans and beneficial insects.
Collapse
Affiliation(s)
- Daniel R Swale
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Department of Entomology, Louisiana State University Agricultural Center, Baton Rouge, LA, 70803, USA
| | - Darren W Engers
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sean R Bollinger
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Aaron Gross
- Department of Entomology and Nematology, University of Florida, Gainesville, FL 32610, USA
| | - Edna Alfaro Inocente
- Department of Entomology, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA
| | - Emily Days
- Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Fariba Kanga
- Department of Entomology, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA
| | - Reed M Johnson
- Department of Entomology, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA
| | - Liu Yang
- Department of Entomology, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA
| | - Jeffrey R Bloomquist
- Department of Entomology and Nematology, University of Florida, Gainesville, FL 32610, USA
| | - Corey R Hopkins
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
| | - Peter M Piermarini
- Department of Entomology, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA
| | - Jerod S Denton
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.,Institute for Global Health, Vanderbilt University, Nashville, TN 37203, USA
| |
Collapse
|
73
|
Dynamic expression of genes encoding subunits of inward rectifier potassium (Kir) channels in the yellow fever mosquito Aedes aegypti. Comp Biochem Physiol B Biochem Mol Biol 2016; 204:35-44. [PMID: 27836744 DOI: 10.1016/j.cbpb.2016.11.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/13/2016] [Accepted: 11/03/2016] [Indexed: 01/06/2023]
Abstract
Inward rectifier potassium (Kir) channels play fundamental roles in neuromuscular, epithelial, and endocrine function in mammals. Recent research in insects suggests that Kir channels play critical roles in the development, immune function, and excretory physiology of fruit flies and/or mosquitoes. Moreover, our group has demonstrated that mosquito Kir channels may serve as valuable targets for the development of novel insecticides. Here we characterize the molecular expression of 5 mRNAs encoding Kir channel subunits in the yellow fever mosquito, Aedes aegypti: Kir1, Kir2A-c, Kir2B, Kir2B', and Kir3. We demonstrate that 1) Kir mRNA expression is dynamic in whole mosquitoes, Malpighian tubules, and the midgut during development from 4th instar larvae to adult females, 2) Kir2B and Kir3 mRNA levels are reduced in 4th instar larvae when reared in water containing an elevated concentration (50mM) of KCl, but not NaCl, and 3) Kir mRNAs are differentially expressed in the Malpighian tubules, midgut, and ovaries within 24h after blood feeding. Furthermore, we provide the first characterization of Kir mRNA expression in the anal papillae of 4th instar larval mosquitoes, which indicates that Kir2A-c is the most abundant. Altogether, the data provide the first comprehensive characterization of Kir mRNA expression in Ae. aegypti and offer insights into the putative physiological roles of Kir subunits in this important disease vector.
Collapse
|
74
|
Formation of new chromatin domains determines pathogenicity of genomic duplications. Nature 2016; 538:265-269. [DOI: 10.1038/nature19800] [Citation(s) in RCA: 455] [Impact Index Per Article: 50.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 08/23/2016] [Indexed: 01/10/2023]
|
75
|
Abstract
Ion channels have emerged as regulators of developmental processes. In model organisms and in people with mutations in ion channels, disruption of ion channel function can affect cell proliferation, cell migration, and craniofacial and limb patterning. Alterations of ion channel function affect morphogenesis in fish, frogs, mammals, and flies, demonstrating that ion channels have conserved roles in developmental processes. One model suggests that ion channels affect proliferation and migration through changes in cell volume. However, ion channels have not explicitly been placed in canonical developmental signaling cascades until recently. This review gives examples of ion channels that influence developmental processes, offers a potential underlying molecular mechanism involving bone morphogenetic protein (BMP) signaling, and finally explores exciting possibilities for manipulating ion channels to influence cell fate for regenerative medicine and to impact disease.
Collapse
Affiliation(s)
- Emily Bates
- Department of Pediatrics, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado 80045;
| |
Collapse
|
76
|
Abstract
Fluid shear stress is an important environmental cue that governs vascular physiology and pathology, but the molecular mechanisms that mediate endothelial responses to flow are only partially understood. Gating of ion channels by flow is one mechanism that may underlie many of the known responses. Here, we review the literature on endothelial ion channels whose activity is modulated by flow with an eye toward identifying important questions for future research.
Collapse
Affiliation(s)
- Kristin A Gerhold
- Department of Internal Medicine (Cardiology), Yale Cardiovascular Research Center, Yale University, New Haven, Connecticut; and
| | - Martin A Schwartz
- Department of Internal Medicine (Cardiology), Yale Cardiovascular Research Center, Yale University, New Haven, Connecticut; and Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, Connecticut
| |
Collapse
|
77
|
Abstract
The central nervous system (CNS) underlies memory, perception, decision-making, and behavior in numerous organisms. However, neural networks have no monopoly on the signaling functions that implement these remarkable algorithms. It is often forgotten that neurons optimized cellular signaling modes that existed long before the CNS appeared during evolution, and were used by somatic cellular networks to orchestrate physiology, embryonic development, and behavior. Many of the key dynamics that enable information processing can, in fact, be implemented by different biological hardware. This is widely exploited by organisms throughout the tree of life. Here, we review data on memory, learning, and other aspects of cognition in a range of models, including single celled organisms, plants, and tissues in animal bodies. We discuss current knowledge of the molecular mechanisms at work in these systems, and suggest several hypotheses for future investigation. The study of cognitive processes implemented in aneural contexts is a fascinating, highly interdisciplinary topic that has many implications for evolution, cell biology, regenerative medicine, computer science, and synthetic bioengineering.
Collapse
Affiliation(s)
- František Baluška
- Department of Plant Cell Biology, IZMB, University of Bonn Bonn, Germany
| | - Michael Levin
- Biology Department, Tufts Center for Regenerative and Developmental Biology, Tufts University Medford, MA, USA
| |
Collapse
|
78
|
Durant F, Lobo D, Hammelman J, Levin M. Physiological controls of large-scale patterning in planarian regeneration: a molecular and computational perspective on growth and form. REGENERATION (OXFORD, ENGLAND) 2016; 3:78-102. [PMID: 27499881 PMCID: PMC4895326 DOI: 10.1002/reg2.54] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/18/2016] [Accepted: 02/22/2016] [Indexed: 12/12/2022]
Abstract
Planaria are complex metazoans that repair damage to their bodies and cease remodeling when a correct anatomy has been achieved. This model system offers a unique opportunity to understand how large-scale anatomical homeostasis emerges from the activities of individual cells. Much progress has been made on the molecular genetics of stem cell activity in planaria. However, recent data also indicate that the global pattern is regulated by physiological circuits composed of ionic and neurotransmitter signaling. Here, we overview the multi-scale problem of understanding pattern regulation in planaria, with specific focus on bioelectric signaling via ion channels and gap junctions (electrical synapses), and computational efforts to extract explanatory models from functional and molecular data on regeneration. We present a perspective that interprets results in this fascinating field using concepts from dynamical systems theory and computational neuroscience. Serving as a tractable nexus between genetic, physiological, and computational approaches to pattern regulation, planarian pattern homeostasis harbors many deep insights for regenerative medicine, evolutionary biology, and engineering.
Collapse
Affiliation(s)
- Fallon Durant
- Department of Biology, Allen Discovery Center at Tufts University, Tufts Center for Regenerative and Developmental BiologyTufts UniversityMA02155USA
| | - Daniel Lobo
- Department of Biological SciencesUniversity of MarylandBaltimore County, 1000 Hilltop CircleBaltimoreMD21250USA
| | - Jennifer Hammelman
- Department of Biology, Allen Discovery Center at Tufts University, Tufts Center for Regenerative and Developmental BiologyTufts UniversityMA02155USA
| | - Michael Levin
- Department of Biology, Allen Discovery Center at Tufts University, Tufts Center for Regenerative and Developmental BiologyTufts UniversityMA02155USA
| |
Collapse
|
79
|
Telocytes in their context with other intercellular communication agents. Semin Cell Dev Biol 2016; 55:9-13. [PMID: 27013113 DOI: 10.1016/j.semcdb.2016.03.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 03/13/2016] [Accepted: 03/15/2016] [Indexed: 11/23/2022]
Abstract
The past decade has borne witness to an explosion in our understanding of the fundamental complexities of intercellular communication. Previously, the field was solely defined by the simple exchange of endocrine, autocrine and epicrine agents. Then it was discovered that cells possess an elaborate system of extracellular vesicles, including exosomes, which carry a vast array of small and large molecules (including many epigenetic agents such as a variety RNAs and DNA), as well as large organelles that modulate almost every aspect of cellular function. In addition, it was thought that electrical communication between cells was limited mainly to neurotransmitters and neuromodulators in the nervous system. Also within the past decade, it was found that - in addition to neurons - most cells (both mammalian and non-mammalian) communicate via elaborate bioelectric systems which modulate many fundamental cellular processes including growth, differentiation, morphogenesis and repair. In the nervous system, volume transmission via the extracellular matrix has been added to the list. Lastly, it was discovered that what had previously been regarded as simple connective cells in most tissues proved to be miniature communication devices now known as telocytes. These unusually long, tenuous and sinuous cells utilize elaborate electrical, chemical and epigenetic mechanisms, including the exchange of exosomes, to integrate many activities within and between nearly all types of cells in tissues and organs. Their interrelationship with neural stem cells and neurogenesis in the context of neurodegenerative disease is just beginning to be explored. This review presents an account of precisely how each of these varied mechanisms are relevant and critical to the understanding of what telocytes are and how they function.
Collapse
|
80
|
Pini J, Rouleau M, Desnuelle C, Sacconi S, Bendahhou S. Modeling Andersen's Syndrome in Human Induced Pluripotent Stem Cells. Stem Cells Dev 2015; 25:151-9. [PMID: 26573604 DOI: 10.1089/scd.2015.0258] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Andersen's syndrome (AS) is a rare disorder characterized by a triad of symptoms: periodic paralysis, cardiac arrhythmia, and bone developmental defects. Most of the patients carry mutations on the inward rectifier potassium channel Kir2.1 encoded by the KCNJ2 gene. kcnj2 knockout mice are lethal at birth preventing, hence, thorough investigations of the physiological and pathophysiological events. We have generated induced pluripotent stem (iPS) cells from healthy as well as from AS patient muscular biopsies using the four-gene cassette required for cellular reprogramming (Oct4, Sox2, Klf4, and c-Myc). The generated AS-iPS cells exhibited the gold standard requirement for iPS cells: expression of genetics and surface pluripotent markers, strong alkaline phosphatase activity, self-renewal, and could be differentiated by the formation of embryoid bodies (EBs) into the three germ layers. Sequencing of the entire coding sequence of the KCNJ2 gene, in AS-iPS cells, revealed that the reprogramming process did not revert the Andersen's syndrome-associated mutation. Moreover, no difference was observed between control and AS-iPS cells in terms of pluripotent markers' expression, self-renewal, and three germ layer differentiation. Interestingly, expression of osteogenic markers are lower in EB-differentiated AS-iPS compared to control iPS cells. Our results showed that the Kir2.1 channel is not important for the reprogramming process and the early step of the development in vitro. However, the osteogenic machinery appears to be hastened in AS-iPS cells, strongly indicating that the generated AS-iPS cells could be a good model to better understand the AS pathophysiology.
Collapse
Affiliation(s)
- Jonathan Pini
- 1 UMR7370 CNRS, LP2M, Labex ICST, Faculté de Médecine, University Nice Sophia Antipolis , Nice, France
| | - Matthieu Rouleau
- 1 UMR7370 CNRS, LP2M, Labex ICST, Faculté de Médecine, University Nice Sophia Antipolis , Nice, France
| | - Claude Desnuelle
- 2 INSERM, U1081, Institute for Research on Cancer and Aging of Nice (IRCAN) , Nice, France .,3 CNRS, UMR 7284, Institute for Research on Cancer and Aging of Nice (IRCAN) , Nice, France .,4 Faculty of Medicine, Neuromuscular Diseases and ALS Specialized Center, University of Nice-Sophia-Antipolis , Nice, France
| | - Sabrina Sacconi
- 2 INSERM, U1081, Institute for Research on Cancer and Aging of Nice (IRCAN) , Nice, France .,3 CNRS, UMR 7284, Institute for Research on Cancer and Aging of Nice (IRCAN) , Nice, France .,4 Faculty of Medicine, Neuromuscular Diseases and ALS Specialized Center, University of Nice-Sophia-Antipolis , Nice, France
| | - Saïd Bendahhou
- 1 UMR7370 CNRS, LP2M, Labex ICST, Faculté de Médecine, University Nice Sophia Antipolis , Nice, France
| |
Collapse
|
81
|
Pai VP, Martyniuk CJ, Echeverri K, Sundelacruz S, Kaplan DL, Levin M. Genome-wide analysis reveals conserved transcriptional responses downstream of resting potential change in Xenopus embryos, axolotl regeneration, and human mesenchymal cell differentiation. ACTA ACUST UNITED AC 2015; 3:3-25. [PMID: 27499876 PMCID: PMC4857752 DOI: 10.1002/reg2.48] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 08/20/2015] [Accepted: 08/25/2015] [Indexed: 12/14/2022]
Abstract
Endogenous bioelectric signaling via changes in cellular resting potential (Vmem) is a key regulator of patterning during regeneration and embryogenesis in numerous model systems. Depolarization of Vmem has been functionally implicated in dedifferentiation, tumorigenesis, anatomical re‐specification, and appendage regeneration. However, no unbiased analyses have been performed to understand genome‐wide transcriptional responses to Vmem change in vivo. Moreover, it is unknown which genes or gene networks represent conserved targets of bioelectrical signaling across different patterning contexts and species. Here, we use microarray analysis to comparatively analyze transcriptional responses to Vmem depolarization. We compare the response of the transcriptome during embryogenesis (Xenopus development), regeneration (axolotl regeneration), and stem cell differentiation (human mesenchymal stem cells in culture) to identify common networks across model species that are associated with depolarization. Both subnetwork enrichment and PANTHER analyses identified a number of key genetic modules as targets of Vmem change, and also revealed important (well‐conserved) commonalities in bioelectric signal transduction, despite highly diverse experimental contexts and species. Depolarization regulates specific transcriptional networks across all three germ layers (ectoderm, mesoderm, and endoderm) such as cell differentiation and apoptosis, and this information will be used for developing mechanistic models of bioelectric regulation of patterning. Moreover, our analysis reveals that Vmem change regulates transcripts related to important disease pathways such as cancer and neurodegeneration, which may represent novel targets for emerging electroceutical therapies.
Collapse
Affiliation(s)
- Vaibhav P Pai
- Biology Department and Center for Regenerative and Developmental Biology Tufts University Medford Massachusetts 02155 USA
| | - Christopher J Martyniuk
- Center for Environmental and Human Toxicology and Department of Physiological Sciences UF Genetics Institute, University of Florida Gainesville Florida 32611 USA
| | - Karen Echeverri
- Department of Genetics, Cell Biology and Development University of Minnesota Minneapolis Minnesota 55455 USA
| | - Sarah Sundelacruz
- Department of Biomedical Engineering Tufts University Medford Massachusetts 02155 USA
| | - David L Kaplan
- Department of Biomedical Engineering Tufts University Medford Massachusetts 02155 USA
| | - Michael Levin
- Biology Department and Center for Regenerative and Developmental Biology Tufts University Medford Massachusetts 02155 USA
| |
Collapse
|
82
|
Pezzulo G, Levin M. Re-membering the body: applications of computational neuroscience to the top-down control of regeneration of limbs and other complex organs. Integr Biol (Camb) 2015; 7:1487-517. [PMID: 26571046 DOI: 10.1039/c5ib00221d] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A major goal of regenerative medicine and bioengineering is the regeneration of complex organs, such as limbs, and the capability to create artificial constructs (so-called biobots) with defined morphologies and robust self-repair capabilities. Developmental biology presents remarkable examples of systems that self-assemble and regenerate complex structures toward their correct shape despite significant perturbations. A fundamental challenge is to translate progress in molecular genetics into control of large-scale organismal anatomy, and the field is still searching for an appropriate theoretical paradigm for facilitating control of pattern homeostasis. However, computational neuroscience provides many examples in which cell networks - brains - store memories (e.g., of geometric configurations, rules, and patterns) and coordinate their activity towards proximal and distant goals. In this Perspective, we propose that programming large-scale morphogenesis requires exploiting the information processing by which cellular structures work toward specific shapes. In non-neural cells, as in the brain, bioelectric signaling implements information processing, decision-making, and memory in regulating pattern and its remodeling. Thus, approaches used in computational neuroscience to understand goal-seeking neural systems offer a toolbox of techniques to model and control regenerative pattern formation. Here, we review recent data on developmental bioelectricity as a regulator of patterning, and propose that target morphology could be encoded within tissues as a kind of memory, using the same molecular mechanisms and algorithms so successfully exploited by the brain. We highlight the next steps of an unconventional research program, which may allow top-down control of growth and form for numerous applications in regenerative medicine and synthetic bioengineering.
Collapse
Affiliation(s)
- G Pezzulo
- Institute of Cognitive Sciences and Technologies, National Research Council, Rome, Italy
| | | |
Collapse
|
83
|
Law R, Levin M. Bioelectric memory: modeling resting potential bistability in amphibian embryos and mammalian cells. Theor Biol Med Model 2015; 12:22. [PMID: 26472354 PMCID: PMC4608135 DOI: 10.1186/s12976-015-0019-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/27/2015] [Indexed: 12/20/2022] Open
Abstract
Background Bioelectric gradients among all cells, not just within excitable nerve and muscle, play instructive roles in developmental and regenerative pattern formation. Plasma membrane resting potential gradients regulate cell behaviors by regulating downstream transcriptional and epigenetic events. Unlike neurons, which fire rapidly and typically return to the same polarized state, developmental bioelectric signaling involves many cell types stably maintaining various levels of resting potential during morphogenetic events. It is important to begin to quantitatively model the stability of bioelectric states in cells, to understand computation and pattern maintenance during regeneration and remodeling. Method To facilitate the analysis of endogenous bioelectric signaling and the exploitation of voltage-based cellular controls in synthetic bioengineering applications, we sought to understand the conditions under which somatic cells can stably maintain distinct resting potential values (a type of state memory). Using the Channelpedia ion channel database, we generated an array of amphibian oocyte and mammalian membrane models for voltage evolution. These models were analyzed and searched, by simulation, for a simple dynamical property, multistability, which forms a type of voltage memory. Results We find that typical mammalian models and amphibian oocyte models exhibit bistability when expressing different ion channel subsets, with either persistent sodium or inward-rectifying potassium, respectively, playing a facilitative role in bistable memory formation. We illustrate this difference using fast sodium channel dynamics for which a comprehensive theory exists, where the same model exhibits bistability under mammalian conditions but not amphibian conditions. In amphibians, potassium channels from the Kv1.x and Kv2.x families tend to disrupt this bistable memory formation. We also identify some common principles under which physiological memory emerges, which suggest specific strategies for implementing memories in bioengineering contexts. Conclusion Our results reveal conditions under which cells can stably maintain one of several resting voltage potential values. These models suggest testable predictions for experiments in developmental bioelectricity, and illustrate how cells can be used as versatile physiological memory elements in synthetic biology, and unconventional computation contexts. Electronic supplementary material The online version of this article (doi:10.1186/s12976-015-0019-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Robert Law
- Department of Neuroscience, Brown University, Box G, Providence, RI, 02912, USA.
| | - Michael Levin
- Department of Biology and Tufts Center for Regenerative and Developmental Biology, Tufts University, 200 Boston Avenue, Medford, MA, 02155, USA.
| |
Collapse
|
84
|
Machado RD, Southgate L, Eichstaedt CA, Aldred MA, Austin ED, Best DH, Chung WK, Benjamin N, Elliott CG, Eyries M, Fischer C, Gräf S, Hinderhofer K, Humbert M, Keiles SB, Loyd JE, Morrell NW, Newman JH, Soubrier F, Trembath RC, Viales RR, Grünig E. Pulmonary Arterial Hypertension: A Current Perspective on Established and Emerging Molecular Genetic Defects. Hum Mutat 2015; 36:1113-27. [PMID: 26387786 DOI: 10.1002/humu.22904] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 09/04/2015] [Indexed: 12/20/2022]
Abstract
Pulmonary arterial hypertension (PAH) is an often fatal disorder resulting from several causes including heterogeneous genetic defects. While mutations in the bone morphogenetic protein receptor type II (BMPR2) gene are the single most common causal factor for hereditary cases, pathogenic mutations have been observed in approximately 25% of idiopathic PAH patients without a prior family history of disease. Additional defects of the transforming growth factor beta pathway have been implicated in disease pathogenesis. Specifically, studies have confirmed activin A receptor type II-like 1 (ACVRL1), endoglin (ENG), and members of the SMAD family as contributing to PAH both with and without associated clinical phenotypes. Most recently, next-generation sequencing has identified novel, rare genetic variation implicated in the PAH disease spectrum. Of importance, several identified genetic factors converge on related pathways and provide significant insight into the development, maintenance, and pathogenetic transformation of the pulmonary vascular bed. Together, these analyses represent the largest comprehensive compilation of BMPR2 and associated genetic risk factors for PAH, comprising known and novel variation. Additionally, with the inclusion of an allelic series of locus-specific variation in BMPR2, these data provide a key resource in data interpretation and development of contemporary therapeutic and diagnostic tools.
Collapse
Affiliation(s)
- Rajiv D Machado
- School of Life Sciences, University of Lincoln, Lincoln, United Kingdom
| | - Laura Southgate
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Division of Genetics & Molecular Medicine, King's College London, London, United Kingdom
| | - Christina A Eichstaedt
- Centre for Pulmonary Hypertension, Thoraxclinic at the University Hospital Heidelberg, Heidelberg, Germany.,Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | | | - Eric D Austin
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - D Hunter Best
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah.,ARUP Institute for Clinical and Experimental Pathology, ARUP Laboratories, Salt Lake City, Utah
| | - Wendy K Chung
- Departments of Pediatrics and Medicine, Columbia University Medical Center, New York, New York
| | - Nicola Benjamin
- Centre for Pulmonary Hypertension, Thoraxclinic at the University Hospital Heidelberg, Heidelberg, Germany
| | - C Gregory Elliott
- Departments of Medicine, Intermountain Medical Center and the University of Utah School of Medicine, Salt Lake City, Utah
| | - Mélanie Eyries
- Unité Mixte de Recherche en Santé (UMR_S 1166), Université Pierre and Marie Curie Université Paris 06 (UPMC) and Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Genetics Department, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.,Institute for Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Christine Fischer
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Stefan Gräf
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom.,Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | | | - Marc Humbert
- Université Paris-Sud, Faculté de Médecine, Paris, France.,Département Hospitalo-Universitaire (DHU) Thorax Innovation (TORINO), Service de Pneumologie, Hôpital Bicêtre, AP-HP, Paris, France.,INSERM UMR_S 999, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Centre Chirurgical Marie Lannelongue, Paris, France
| | - Steven B Keiles
- Quest Diagnostics, Action from Insight, San Juan Capistrano, California
| | - James E Loyd
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom.,Addenbrooke's & Papworth Hospitals, Cambridge, United Kingdom
| | - John H Newman
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Florent Soubrier
- Unité Mixte de Recherche en Santé (UMR_S 1166), Université Pierre and Marie Curie Université Paris 06 (UPMC) and Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France.,Genetics Department, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.,Institute for Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Richard C Trembath
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Rebecca Rodríguez Viales
- Centre for Pulmonary Hypertension, Thoraxclinic at the University Hospital Heidelberg, Heidelberg, Germany.,Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Ekkehard Grünig
- Centre for Pulmonary Hypertension, Thoraxclinic at the University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
85
|
Villanueva S, Burgos J, López-Cayuqueo KI, Lai KMV, Valenzuela DM, Cid LP, Sepúlveda FV. Cleft Palate, Moderate Lung Developmental Retardation and Early Postnatal Lethality in Mice Deficient in the Kir7.1 Inwardly Rectifying K+ Channel. PLoS One 2015; 10:e0139284. [PMID: 26402555 PMCID: PMC4581704 DOI: 10.1371/journal.pone.0139284] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 09/09/2015] [Indexed: 12/15/2022] Open
Abstract
Kir7.1 is an inwardly rectifying K+ channel of the Kir superfamily encoded by the kcnj13 gene. Kir7.1 is present in epithelial tissues where it colocalizes with the Na+/K+-pump probably serving to recycle K+ taken up by the pump. Human mutations affecting Kir7.1 are associated with retinal degeneration diseases. We generated a mouse lacking Kir7.1 by ablation of the Kcnj13 gene. Homozygous mutant null mice die hours after birth and show cleft palate and moderate retardation in lung development. Kir7.1 is expressed in the epithelium covering the palatal processes at the time at which palate sealing takes place and our results suggest it might play an essential role in late palatogenesis. Our work also reveals a second unexpected role in the development and the physiology of the respiratory system, where Kir7.1 is expressed in epithelial cells all along the respiratory tree.
Collapse
Affiliation(s)
| | - Johanna Burgos
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Doctorado en Ciencias Veterinarias de la Universidad Austral de Chile, Valdivia, Chile
| | | | - Ka-Man Venus Lai
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, United States of America
| | - David M. Valenzuela
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, United States of America
| | - L. Pablo Cid
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| | | |
Collapse
|
86
|
Wu Y, Baum M, Huang CL, Rodan AR. Two inwardly rectifying potassium channels, Irk1 and Irk2, play redundant roles in Drosophila renal tubule function. Am J Physiol Regul Integr Comp Physiol 2015. [PMID: 26224687 DOI: 10.1152/ajpregu.00148.2015] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Inwardly rectifying potassium channels play essential roles in renal physiology across phyla. Barium-sensitive K(+) conductances are found on the basolateral membrane of a variety of insect Malpighian (renal) tubules, including Drosophila melanogaster. We found that barium decreases the lumen-positive transepithelial potential difference in isolated perfused Drosophila tubules and decreases fluid secretion and transepithelial K(+) flux. In those insect species in which it has been studied, transcripts from multiple genes encoding inwardly rectifying K(+) channels are expressed in the renal (Malpighian) tubule. In Drosophila melanogaster, this includes transcripts of the Irk1, Irk2, and Irk3 genes. The role of each of these gene products in renal tubule function is unknown. We found that simultaneous knockdown of Irk1 and Irk2 in the principal cell of the fly tubule decreases transepithelial K(+) flux, with no additive effect of Irk3 knockdown, and decreases barium sensitivity of transepithelial K(+) flux by ∼50%. Knockdown of any of the three inwardly rectifying K(+) channels individually has no effect, nor does knocking down Irk3 simultaneously with Irk1 or Irk2. Irk1/Irk2 principal cell double-knockdown tubules remain sensitive to the kaliuretic effect of cAMP. Inhibition of the Na(+)/K(+)-ATPase with ouabain and Irk1/Irk2 double knockdown have additive effects on K(+) flux, and 75% of transepithelial K(+) transport is due to Irk1/Irk2 or ouabain-sensitive pathways. In conclusion, Irk1 and Irk2 play redundant roles in transepithelial ion transport in the Drosophila melanogaster renal tubule and are additive to Na(+)/K(+)-ATPase-dependent pathways.
Collapse
Affiliation(s)
- Yipin Wu
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas; and
| | - Michel Baum
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas; and Department of Pediatrics, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chou-Long Huang
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas; and
| | - Aylin R Rodan
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas; and
| |
Collapse
|
87
|
Levin M. Molecular bioelectricity: how endogenous voltage potentials control cell behavior and instruct pattern regulation in vivo. Mol Biol Cell 2015; 25:3835-50. [PMID: 25425556 PMCID: PMC4244194 DOI: 10.1091/mbc.e13-12-0708] [Citation(s) in RCA: 253] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In addition to biochemical gradients and transcriptional networks, cell behavior is regulated by endogenous bioelectrical cues originating in the activity of ion channels and pumps, operating in a wide variety of cell types. Instructive signals mediated by changes in resting potential control proliferation, differentiation, cell shape, and apoptosis of stem, progenitor, and somatic cells. Of importance, however, cells are regulated not only by their own Vmem but also by the Vmem of their neighbors, forming networks via electrical synapses known as gap junctions. Spatiotemporal changes in Vmem distribution among nonneural somatic tissues regulate pattern formation and serve as signals that trigger limb regeneration, induce eye formation, set polarity of whole-body anatomical axes, and orchestrate craniofacial patterning. New tools for tracking and functionally altering Vmem gradients in vivo have identified novel roles for bioelectrical signaling and revealed the molecular pathways by which Vmem changes are transduced into cascades of downstream gene expression. Because channels and gap junctions are gated posttranslationally, bioelectrical networks have their own characteristic dynamics that do not reduce to molecular profiling of channel expression (although they couple functionally to transcriptional networks). The recent data provide an exciting opportunity to crack the bioelectric code, and learn to program cellular activity at the level of organs, not only cell types. The understanding of how patterning information is encoded in bioelectrical networks, which may require concepts from computational neuroscience, will have transformative implications for embryogenesis, regeneration, cancer, and synthetic bioengineering.
Collapse
Affiliation(s)
- Michael Levin
- Biology Department, Center for Regenerative and Developmental Biology, Tufts University, Medford, MA 02155-4243
| |
Collapse
|
88
|
Pai VP, Lemire JM, Paré JF, Lin G, Chen Y, Levin M. Endogenous gradients of resting potential instructively pattern embryonic neural tissue via Notch signaling and regulation of proliferation. J Neurosci 2015; 35:4366-85. [PMID: 25762681 PMCID: PMC4355204 DOI: 10.1523/jneurosci.1877-14.2015] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 12/21/2014] [Accepted: 01/14/2015] [Indexed: 12/26/2022] Open
Abstract
Biophysical forces play important roles throughout embryogenesis, but the roles of spatial differences in cellular resting potentials during large-scale brain morphogenesis remain unknown. Here, we implicate endogenous bioelectricity as an instructive factor during brain patterning in Xenopus laevis. Early frog embryos exhibit a characteristic hyperpolarization of cells lining the neural tube; disruption of this spatial gradient of the transmembrane potential (Vmem) diminishes or eliminates the expression of early brain markers, and causes anatomical mispatterning of the brain, including absent or malformed regions. This effect is mediated by voltage-gated calcium signaling and gap-junctional communication. In addition to cell-autonomous effects, we show that hyperpolarization of transmembrane potential (Vmem) in ventral cells outside the brain induces upregulation of neural cell proliferation at long range. Misexpression of the constitutively active form of Notch, a suppressor of neural induction, impairs the normal hyperpolarization pattern and neural patterning; forced hyperpolarization by misexpression of specific ion channels rescues brain defects induced by activated Notch signaling. Strikingly, hyperpolarizing posterior or ventral cells induces the production of ectopic neural tissue considerably outside the neural field. The hyperpolarization signal also synergizes with canonical reprogramming factors (POU and HB4), directing undifferentiated cells toward neural fate in vivo. These data identify a new functional role for bioelectric signaling in brain patterning, reveal interactions between Vmem and key biochemical pathways (Notch and Ca(2+) signaling) as the molecular mechanism by which spatial differences of Vmem regulate organogenesis of the vertebrate brain, and suggest voltage modulation as a tractable strategy for intervention in certain classes of birth defects.
Collapse
Affiliation(s)
- Vaibhav P Pai
- Biology Department, Center for Regenerative and Developmental Biology, Tufts University, Medford, Massachusetts 02155-4243 and
| | - Joan M Lemire
- Biology Department, Center for Regenerative and Developmental Biology, Tufts University, Medford, Massachusetts 02155-4243 and
| | - Jean-François Paré
- Biology Department, Center for Regenerative and Developmental Biology, Tufts University, Medford, Massachusetts 02155-4243 and
| | - Gufa Lin
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota 55455
| | - Ying Chen
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota 55455
| | - Michael Levin
- Biology Department, Center for Regenerative and Developmental Biology, Tufts University, Medford, Massachusetts 02155-4243 and
| |
Collapse
|
89
|
Cervera J, Manzanares JA, Mafe S. Electrical coupling in ensembles of nonexcitable cells: modeling the spatial map of single cell potentials. J Phys Chem B 2015; 119:2968-78. [PMID: 25622192 DOI: 10.1021/jp512900x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
We analyze the coupling of model nonexcitable (non-neural) cells assuming that the cell membrane potential is the basic individual property. We obtain this potential on the basis of the inward and outward rectifying voltage-gated channels characteristic of cell membranes. We concentrate on the electrical coupling of a cell ensemble rather than on the biochemical and mechanical characteristics of the individual cells, obtain the map of single cell potentials using simple assumptions, and suggest procedures to collectively modify this spatial map. The response of the cell ensemble to an external perturbation and the consequences of cell isolation, heterogeneity, and ensemble size are also analyzed. The results suggest that simple coupling mechanisms can be significant for the biophysical chemistry of model biomolecular ensembles. In particular, the spatiotemporal map of single cell potentials should be relevant for the uptake and distribution of charged nanoparticles over model cell ensembles and the collective properties of droplet networks incorporating protein ion channels inserted in lipid bilayers.
Collapse
Affiliation(s)
- Javier Cervera
- Departament de Termodinàmica, Universitat de València , E-46100 Burjassot, Spain
| | | | | |
Collapse
|
90
|
Krüger J, Bohrmann J. Bioelectric patterning during oogenesis: stage-specific distribution of membrane potentials, intracellular pH and ion-transport mechanisms in Drosophila ovarian follicles. BMC DEVELOPMENTAL BIOLOGY 2015; 15:1. [PMID: 25591552 PMCID: PMC4302609 DOI: 10.1186/s12861-015-0051-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 01/05/2015] [Indexed: 01/01/2023]
Abstract
Background Bioelectric phenomena have been found to exert influence on various developmental and regenerative processes. Little is known about their possible functions and the cellular mechanisms by which they might act during Drosophila oogenesis. In developing follicles, characteristic extracellular current patterns and membrane-potential changes in oocyte and nurse cells have been observed that partly depend on the exchange of protons, potassium ions and sodium ions. These bioelectric properties have been supposed to be related to various processes during oogenesis, e. g. pH-regulation, osmoregulation, cell communication, cell migration, cell proliferation, cell death, vitellogenesis and follicle growth. Analysing in detail the spatial distribution and activity of the relevant ion-transport mechanisms is expected to elucidate the roles that bioelectric phenomena play during oogenesis. Results To obtain an overview of bioelectric patterning along the longitudinal and transversal axes of the developing follicle, the spatial distributions of membrane potentials (Vmem), intracellular pH (pHi) and various membrane-channel proteins were studied systematically using fluorescent indicators, fluorescent inhibitors and antisera. During mid-vitellogenic stages 9 to 10B, characteristic, stage-specific Vmem-patterns in the follicle-cell epithelium as well as anteroposterior pHi-gradients in follicle cells and nurse cells were observed. Corresponding distribution patterns of proton pumps (V-ATPases), voltage-dependent L-type Ca2+-channels, amiloride-sensitive Na+-channels and Na+,H+-exchangers (NHE) and gap-junction proteins (innexin 3) were detected. In particular, six morphologically distinguishable follicle-cell types are characterized on the bioelectric level by differences concerning Vmem and pHi as well as specific compositions of ion channels and carriers. Striking similarities between Vmem-patterns and activity patterns of voltage-dependent Ca2+-channels were found, suggesting a mechanism for transducing bioelectric signals into cellular responses. Moreover, gradients of electrical potential and pH were observed within single cells. Conclusions Our data suggest that spatial patterning of Vmem, pHi and specific membrane-channel proteins results in bioelectric signals that are supposed to play important roles during oogenesis, e. g. by influencing spatial coordinates, regulating migration processes or modifying the cytoskeletal organization. Characteristic stage-specific changes of bioelectric activity in specialized cell types are correlated with various developmental processes.
Collapse
Affiliation(s)
- Julia Krüger
- RWTH Aachen University, Institut für Biologie II, Abt. Zoologie und Humanbiologie, Worringerweg 3, 52056, Aachen, Germany.
| | - Johannes Bohrmann
- RWTH Aachen University, Institut für Biologie II, Abt. Zoologie und Humanbiologie, Worringerweg 3, 52056, Aachen, Germany.
| |
Collapse
|
91
|
Levin M. Endogenous bioelectrical networks store non-genetic patterning information during development and regeneration. J Physiol 2015; 592:2295-305. [PMID: 24882814 DOI: 10.1113/jphysiol.2014.271940] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Pattern formation, as occurs during embryogenesis or regeneration, is the crucial link between genotype and the functions upon which selection operates. Even cancer and aging can be seen as challenges to the continuous physiological processes that orchestrate individual cell activities toward the anatomical needs of an organism. Thus, the origin and maintenance of complex biological shape is a fundamental question for cell, developmental, and evolutionary biology, as well as for biomedicine. It has long been recognized that slow bioelectrical gradients can control cell behaviors and morphogenesis. Here, I review recent molecular data that implicate endogenous spatio-temporal patterns of resting potentials among non-excitable cells as instructive cues in embryogenesis, regeneration, and cancer. Functional data have implicated gradients of resting potential in processes such as limb regeneration, eye induction, craniofacial patterning, and head-tail polarity, as well as in metastatic transformation and tumorigenesis. The genome is tightly linked to bioelectric signaling, via ion channel proteins that shape the gradients, downstream genes whose transcription is regulated by voltage, and transduction machinery that converts changes in bioelectric state to second-messenger cascades. However, the data clearly indicate that bioelectric signaling is an autonomous layer of control not reducible to a biochemical or genetic account of cell state. The real-time dynamics of bioelectric communication among cells are not fully captured by transcriptomic or proteomic analyses, and the necessary-and-sufficient triggers for specific changes in growth and form can be physiological states, while the underlying gene loci are free to diverge. The next steps in this exciting new field include the development of novel conceptual tools for understanding the anatomical semantics encoded in non-neural bioelectrical networks, and of improved biophysical tools for reading and writing electrical state information into somatic tissues. Cracking the bioelectric code will have transformative implications for developmental biology, regenerative medicine, and synthetic bioengineering.
Collapse
Affiliation(s)
- Michael Levin
- Biology Department, Center for Regenerative and Developmental Biology, Tufts University, Medford, MA, USA
| |
Collapse
|
92
|
Raphemot R, Rouhier MF, Swale DR, Days E, Weaver CD, Lovell KM, Konkel LC, Engers DW, Bollinger SF, Hopkins C, Piermarini PM, Denton JS. Discovery and characterization of a potent and selective inhibitor of Aedes aegypti inward rectifier potassium channels. PLoS One 2014; 9:e110772. [PMID: 25375326 PMCID: PMC4222822 DOI: 10.1371/journal.pone.0110772] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 09/16/2014] [Indexed: 12/12/2022] Open
Abstract
Vector-borne diseases such as dengue fever and malaria, which are transmitted by infected female mosquitoes, affect nearly half of the world's population. The emergence of insecticide-resistant mosquito populations is reducing the effectiveness of conventional insecticides and threatening current vector control strategies, which has created an urgent need to identify new molecular targets against which novel classes of insecticides can be developed. We previously demonstrated that small molecule inhibitors of mammalian Kir channels represent promising chemicals for new mosquitocide development. In this study, high-throughput screening of approximately 30,000 chemically diverse small-molecules was employed to discover potent and selective inhibitors of Aedes aegypti Kir1 (AeKir1) channels heterologously expressed in HEK293 cells. Of 283 confirmed screening ‘hits’, the small-molecule inhibitor VU625 was selected for lead optimization and in vivo studies based on its potency and selectivity toward AeKir1, and tractability for medicinal chemistry. In patch clamp electrophysiology experiments of HEK293 cells, VU625 inhibits AeKir1 with an IC50 value of 96.8 nM, making VU625 the most potent inhibitor of AeKir1 described to date. Furthermore, electrophysiology experiments in Xenopus oocytes revealed that VU625 is a weak inhibitor of AeKir2B. Surprisingly, injection of VU625 failed to elicit significant effects on mosquito behavior, urine excretion, or survival. However, when co-injected with probenecid, VU625 inhibited the excretory capacity of mosquitoes and was toxic, suggesting that the compound is a substrate of organic anion and/or ATP-binding cassette (ABC) transporters. The dose-toxicity relationship of VU625 (when co-injected with probenecid) is biphasic, which is consistent with the molecule inhibiting both AeKir1 and AeKir2B with different potencies. This study demonstrates proof-of-concept that potent and highly selective inhibitors of mosquito Kir channels can be developed using conventional drug discovery approaches. Furthermore, it reinforces the notion that the physical and chemical properties that determine a compound's bioavailability in vivo will be critical in determining the efficacy of Kir channel inhibitors as insecticides.
Collapse
Affiliation(s)
- Rene Raphemot
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, United States of America
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Matthew F. Rouhier
- Department of Entomology, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States of America
| | - Daniel R. Swale
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Emily Days
- Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - C. David Weaver
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Kimberly M. Lovell
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- Department of Chemistry, Vanderbilt University School of Medicine, Nashville TN, United States of America
| | - Leah C. Konkel
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- Department of Chemistry, Vanderbilt University School of Medicine, Nashville TN, United States of America
| | - Darren W. Engers
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- Department of Chemistry, Vanderbilt University School of Medicine, Nashville TN, United States of America
| | - Sean F. Bollinger
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- Department of Chemistry, Vanderbilt University School of Medicine, Nashville TN, United States of America
| | - Corey Hopkins
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- Institute for Global Health, Vanderbilt University, Nashville, TN, United States of America
- Department of Chemistry, Vanderbilt University School of Medicine, Nashville TN, United States of America
| | - Peter M. Piermarini
- Department of Entomology, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States of America
- * E-mail: (PMP); (JSD)
| | - Jerod S. Denton
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, United States of America
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- Institute for Global Health, Vanderbilt University, Nashville, TN, United States of America
- * E-mail: (PMP); (JSD)
| |
Collapse
|
93
|
Cervera J, Alcaraz A, Mafe S. Membrane potential bistability in nonexcitable cells as described by inward and outward voltage-gated ion channels. J Phys Chem B 2014; 118:12444-50. [PMID: 25286866 DOI: 10.1021/jp508304h] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The membrane potential of nonexcitable cells, defined as the electrical potential difference between the cell cytoplasm and the extracellular environment when the current is zero, is controlled by the individual electrical conductance of different ion channels. In particular, inward- and outward-rectifying voltage-gated channels are crucial for cell hyperpolarization/depolarization processes, being amenable to direct physical study. High (in absolute value) negative membrane potentials are characteristic of terminally differentiated cells, while low membrane potentials are found in relatively depolarized, more plastic cells (e.g., stem, embryonic, and cancer cells). We study theoretically the hyperpolarized and depolarized values of the membrane potential, as well as the possibility to obtain a bistability behavior, using simplified models for the ion channels that regulate this potential. The bistability regions, which are defined in the multidimensional state space determining the cell state, can be relevant for the understanding of the different model cell states and the transitions between them, which are triggered by changes in the external environment.
Collapse
Affiliation(s)
- Javier Cervera
- Departament de Termodinàmica, Universitat de València , E-46100 Burjassot, Spain
| | | | | |
Collapse
|
94
|
Sacco S, Giuliano S, Sacconi S, Desnuelle C, Barhanin J, Amri EZ, Bendahhou S. The inward rectifier potassium channel Kir2.1 is required for osteoblastogenesis. Hum Mol Genet 2014; 24:471-9. [PMID: 25205110 DOI: 10.1093/hmg/ddu462] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Andersen's syndrome (AS) is a rare and dominantly inherited pathology, linked to the inwardly rectifying potassium channel Kir2.1. AS patients exhibit a triad of symptoms that include periodic paralysis, cardiac dysrhythmia and bone malformations. Some progress has been made in understanding the contribution of the Kir2.1 channel to skeletal and cardiac muscle dysfunctions, but its role in bone morphogenesis remains unclear. We isolated myoblast precursors from muscle biopsies of healthy individuals and typical AS patients with dysmorphic features. Myoblast cultures underwent osteogenic differentiation that led to extracellular matrix mineralization. Osteoblastogenesis was monitored through the activity of alkaline phosphatase, and through the hydroxyapatite formation using Alizarin Red and Von Kossa staining techniques. Patch-clamp recordings revealed the presence of an inwardly rectifying current in healthy cells that was absent in AS osteoblasts, showing the dominant-negative effect of the Kir2.1 mutant allele in osteoblasts. We also found that while control cells actively synthesize hydroxyapatite, AS osteoblasts are unable to efficiently form any extracellular matrix. To further demonstrate the role of the Kir2.1 channels during the osteogenesis, we inhibited Kir2.1 channel activity in healthy patient cells by applying extracellular Ba(2+) or using adenoviruses carrying mutant Kir2.1 channels. In both cases, cells were no longer able to produce extracellular matrixes. Moreover, osteogenic activity of AS osteoblasts was restored by rescue experiments, via wild-type Kir2.1 channel overexpression. These observations provide a proof that normal Kir2.1 channel function is essential during osteoblastogenesis.
Collapse
Affiliation(s)
- Sonia Sacco
- University Nice Sophia Antipolis, Nice, France
| | - Serena Giuliano
- University Nice Sophia Antipolis, Nice, France Laboratory of Excellence Ion Channel Science and Therapeutics, LP2M, UMR 7370 CNRS, Nice, France Faculté de Médecine, Nice, France
| | - Sabrina Sacconi
- University Nice Sophia Antipolis, Nice, France Neuromuscular Disease Specialized Center, Nice University Hospital, Nice, France IRCAN, UMR 7248 CNRS, INSERM U1081, Nice, France and
| | - Claude Desnuelle
- University Nice Sophia Antipolis, Nice, France Neuromuscular Disease Specialized Center, Nice University Hospital, Nice, France IRCAN, UMR 7248 CNRS, INSERM U1081, Nice, France and
| | - Jacques Barhanin
- University Nice Sophia Antipolis, Nice, France Laboratory of Excellence Ion Channel Science and Therapeutics, LP2M, UMR 7370 CNRS, Nice, France Faculté de Médecine, Nice, France
| | - Ez-zoubir Amri
- University Nice Sophia Antipolis, Nice, France Faculté de Médecine, Nice, France IBV, UMR 7277 CNRS, INSERM U1091, Nice, France
| | - Saïd Bendahhou
- University Nice Sophia Antipolis, Nice, France Laboratory of Excellence Ion Channel Science and Therapeutics, LP2M, UMR 7370 CNRS, Nice, France Faculté de Médecine, Nice, France
| |
Collapse
|
95
|
Mustard J, Levin M. Bioelectrical Mechanisms for Programming Growth and Form: Taming Physiological Networks for Soft Body Robotics. Soft Robot 2014. [DOI: 10.1089/soro.2014.0011] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Jessica Mustard
- Department of Biology and Center for Regenerative and Developmental Biology, Tufts University, Medford, Massachusetts
| | - Michael Levin
- Department of Biology and Center for Regenerative and Developmental Biology, Tufts University, Medford, Massachusetts
| |
Collapse
|
96
|
Raphemot R, Estévez-Lao TY, Rouhier MF, Piermarini PM, Denton JS, Hillyer JF. Molecular and functional characterization of Anopheles gambiae inward rectifier potassium (Kir1) channels: a novel role in egg production. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2014; 51:10-9. [PMID: 24855023 PMCID: PMC4121989 DOI: 10.1016/j.ibmb.2014.05.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 05/03/2014] [Accepted: 05/10/2014] [Indexed: 05/21/2023]
Abstract
Inward rectifier potassium (Kir) channels play essential roles in regulating diverse physiological processes. Although Kir channels are encoded in mosquito genomes, their functions remain largely unknown. In this study, we identified the members of the Anopheles gambiae Kir gene family and began to investigate their function. Notably, we sequenced the A. gambiae Kir1 (AgKir1) gene and showed that it encodes all the canonical features of a Kir channel: an ion pore that is composed of a pore helix and a selectivity filter, two transmembrane domains that flank the ion pore, and the so-called G-loop. Heterologous expression of AgKir1 in Xenopus oocytes revealed that this gene encodes a functional, barium-sensitive Kir channel. Quantitative RT-PCR experiments then showed that relative AgKir1 mRNA levels are highest in the pupal stage, and that AgKir1 mRNA is enriched in the adult ovaries. Gene silencing of AgKir1 by RNA interference did not affect the survival of female mosquitoes following a blood meal, but decreased their egg output. These data provide evidence for a new role of Kir channels in mosquito fecundity, and further validates them as promising molecular targets for the development of a new class of mosquitocides to be used in vector control.
Collapse
Affiliation(s)
- Rene Raphemot
- Department of Anesthesiology, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Tania Y Estévez-Lao
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA
| | - Matthew F Rouhier
- Department of Entomology, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA
| | - Peter M Piermarini
- Department of Entomology, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA
| | - Jerod S Denton
- Department of Anesthesiology, Vanderbilt University, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA; Institute for Global Health, Vanderbilt University, Nashville, TN 37232, USA.
| | - Julián F Hillyer
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA; Institute for Global Health, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
97
|
Rouhier MF, Piermarini PM. Identification of life-stage and tissue-specific splice variants of an inward rectifying potassium (Kir) channel in the yellow fever mosquito Aedes aegypti. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2014; 48:91-99. [PMID: 24657620 DOI: 10.1016/j.ibmb.2014.03.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 03/05/2014] [Accepted: 03/06/2014] [Indexed: 06/03/2023]
Abstract
Inward-rectifier potassium (Kir) channels play key roles in nerve, muscle, and epithelial cells in mammals, but their physiological roles in insects remain to be determined. The yellow fever mosquito (Aedes aegypti) possesses five different genes encoding Kir channel subunits: Kir1, Kir2A, Kir2B, Kir2B', and Kir3. We have recently cloned and characterized the Kir1, Kir2B, and Kir3 cDNAs in the renal (Malpighian) tubules of adult female Ae. aegypti. Here we characterize the expression of the Kir2A gene in Ae. aegypti, which was not abundantly expressed in Malpighian tubules. We find that the 1) Kir2A gene is expressed primarily in the midgut and hindgut of adult female mosquitoes, and 2) Kir2A mRNAs are alternatively spliced into three distinct variants (Kir2A-a, -b, and -c). The deduced Kir2A proteins from these splice forms share a completely conserved transmembrane domain (a pore-forming domain flanked by two transmembrane-spanning segments), but possess novel NH2-terminal and/or COOH-terminal domains. Semi-quantitative RT-PCR analyses indicate that the splice variants exhibit both developmental- and tissue-specific expression. Lastly, we provide insights into the conservation of alternative splicing among the Kir2A genes of dipterans, which may add molecular diversity that compensates for the relatively limited number of Kir channel genes in insects compared to mammals.
Collapse
Affiliation(s)
- Matthew F Rouhier
- Department of Entomology, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA
| | - Peter M Piermarini
- Department of Entomology, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA.
| |
Collapse
|
98
|
Perathoner S, Daane JM, Henrion U, Seebohm G, Higdon CW, Johnson SL, Nüsslein-Volhard C, Harris MP. Bioelectric signaling regulates size in zebrafish fins. PLoS Genet 2014; 10:e1004080. [PMID: 24453984 PMCID: PMC3894163 DOI: 10.1371/journal.pgen.1004080] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 11/19/2013] [Indexed: 12/22/2022] Open
Abstract
The scaling relationship between the size of an appendage or organ and that of the body as a whole is tightly regulated during animal development. If a structure grows at a different rate than the rest of the body, this process is termed allometric growth. The zebrafish another longfin (alf) mutant shows allometric growth resulting in proportionally enlarged fins and barbels. We took advantage of this mutant to study the regulation of size in vertebrates. Here, we show that alf mutants carry gain-of-function mutations in kcnk5b, a gene encoding a two-pore domain potassium (K+) channel. Electrophysiological analysis in Xenopus oocytes reveals that these mutations cause an increase in K+ conductance of the channel and lead to hyperpolarization of the cell. Further, somatic transgenesis experiments indicate that kcnk5b acts locally within the mesenchyme of fins and barbels to specify appendage size. Finally, we show that the channel requires the ability to conduct K+ ions to increase the size of these structures. Our results provide evidence for a role of bioelectric signaling through K+ channels in the regulation of allometric scaling and coordination of growth in the zebrafish. The proportions of an animal can change during its lifetime. This often occurs through the phenomenon of relative growth, or allometry. In humans, for example, the head grows at a lower rate than the body resulting in a change in proportion between children and adults. The regulation of size and proportion is not well understood. We investigated fin growth in zebrafish as a model to understand this phenomenon. The mutant another longfin develops proportionally larger fins and barbels. Here, we show that another longfin mutants carry a mutation in kcnk5b, a gene coding for a potassium channel. Potassium channels control the electrical potential of cells and are known to regulate processes such as heart beat, neurotransmission and hormone secretion. We demonstrate that increased potassium channel activity can drive changes in growth in the zebrafish. Clonal analysis reveals that the channel acts directly in the fins and barbels to increase their size in a coordinated manner. Our work shows that potassium channels are involved in the determination of appendage size in zebrafish and suggests consistent with recent studies, an instructive role for bioelectrical signaling in development.
Collapse
Affiliation(s)
- Simon Perathoner
- Max Planck Institute for Developmental Biology, Tübingen, Germany
- * E-mail: (SP); (CNV)
| | - Jacob M. Daane
- Orthopedic Research Laboratories, Boston Children's Hospital; Department of Genetics, Harvard Medical School, Enders, Massachusetts, United States of America
| | - Ulrike Henrion
- Abteilung Myozelluläre Elektrophysiologie, Institut für Genetik von Herzerkrankungen, Universitätsklinikum Münster, Albert-Schweizer-Campus 1, Münster, Germany
| | - Guiscard Seebohm
- Abteilung Myozelluläre Elektrophysiologie, Institut für Genetik von Herzerkrankungen, Universitätsklinikum Münster, Albert-Schweizer-Campus 1, Münster, Germany
| | - Charles W. Higdon
- Department of Genetics, Washington University Medical School, St. Louis, Missouri, United States of America
| | - Stephen L. Johnson
- Department of Genetics, Washington University Medical School, St. Louis, Missouri, United States of America
| | | | - Matthew P. Harris
- Orthopedic Research Laboratories, Boston Children's Hospital; Department of Genetics, Harvard Medical School, Enders, Massachusetts, United States of America
| |
Collapse
|
99
|
Tian C, Zhu R, Zhu L, Qiu T, Cao Z, Kang T. Potassium Channels: Structures, Diseases, and Modulators. Chem Biol Drug Des 2013; 83:1-26. [DOI: 10.1111/cbdd.12237] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Chuan Tian
- School of Life Sciences and Technology; Tongji University; Shanghai 200092 China
- School of Pharmacy; Liaoning University of Traditional Chinese Medicine; Dalian Liaoning 116600 China
| | - Ruixin Zhu
- School of Life Sciences and Technology; Tongji University; Shanghai 200092 China
| | - Lixin Zhu
- Department of Pediatrics; Digestive Diseases and Nutrition Center; The State University of New York at Buffalo; Buffalo NY 14226 USA
| | - Tianyi Qiu
- School of Life Sciences and Technology; Tongji University; Shanghai 200092 China
| | - Zhiwei Cao
- School of Life Sciences and Technology; Tongji University; Shanghai 200092 China
| | - Tingguo Kang
- School of Pharmacy; Liaoning University of Traditional Chinese Medicine; Dalian Liaoning 116600 China
| |
Collapse
|
100
|
|