1
|
Prelic S, Keesey IW, Lavista-Llanos S, Hansson BS, Wicher D. Innexin expression and localization in the Drosophila antenna indicate gap junction or hemichannel involvement in antennal chemosensory sensilla. Cell Tissue Res 2024; 398:35-62. [PMID: 39174822 PMCID: PMC11424723 DOI: 10.1007/s00441-024-03909-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 07/25/2024] [Indexed: 08/24/2024]
Abstract
Odor detection in insects is largely mediated by structures on antennae called sensilla, which feature a strongly conserved architecture and repertoire of olfactory sensory neurons (OSNs) and various support cell types. In Drosophila, OSNs are tightly apposed to supporting cells, whose connection with neurons and functional roles in odor detection remain unclear. Coupling mechanisms between these neuronal and non-neuronal cell types have been suggested based on morphological observations, concomitant physiological activity during odor stimulation, and known interactions that occur in other chemosensory systems. For instance, it is not known whether cell-cell coupling via gap junctions between OSNs and neighboring cells exists, or whether hemichannels interconnect cellular and extracellular sensillum compartments. Here, we show that innexins, which form hemichannels and gap junctions in invertebrates, are abundantly expressed in adult drosophilid antennae. By surveying antennal transcriptomes and performing various immunohistochemical stainings in antennal tissues, we discover innexin-specific patterns of expression and localization, with a majority of innexins strongly localizing to glial and non-neuronal cells, likely support and epithelial cells. Finally, by injecting gap junction-permeable dye into a pre-identified sensillum, we observe no dye coupling between neuronal and non-neuronal cells. Together with evidence of non-neuronal innexin localization, we conclude that innexins likely do not conjoin neurons to support cells, but that junctions and hemichannels may instead couple support cells among each other or to their shared sensillum lymph to achieve synchronous activity. We discuss how coupling of sensillum microenvironments or compartments may potentially contribute to facilitate chemosensory functions of odor sensing and sensillum homeostasis.
Collapse
Affiliation(s)
- Sinisa Prelic
- Dept. Evolutionary Neuroethology, Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Ian W Keesey
- Dept. Evolutionary Neuroethology, Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Sofia Lavista-Llanos
- Dept. Evolutionary Neuroethology, Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Bill S Hansson
- Dept. Evolutionary Neuroethology, Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Dieter Wicher
- Dept. Evolutionary Neuroethology, Max Planck Institute for Chemical Ecology, Jena, Germany.
| |
Collapse
|
2
|
Meng JH, Huang YB, Long J, Cai QC, Qiao X, Zhang QL, Zhang LD, Yan X, Jing R, Liu XS, Zhou SJ, Yuan YS, Yin-Chen Ma, Zhou LX, Peng NN, Li XC, Cai CH, Tang HM, Martins AF, Jiang JX, Kai-Jun Luo. Innexin hemichannel activation by Microplitis bicoloratus ecSOD monopolymer reduces ROS. iScience 2024; 27:109469. [PMID: 38577101 PMCID: PMC10993139 DOI: 10.1016/j.isci.2024.109469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 01/31/2024] [Accepted: 03/07/2024] [Indexed: 04/06/2024] Open
Abstract
The extracellular superoxide dismutases (ecSODs) secreted by Microplitis bicoloratus reduce the reactive oxygen species (ROS) stimulated by the Microplitis bicoloratus bracovirus. Here, we demonstrate that the bacterial transferase hexapeptide (hexapep) motif and bacterial-immunoglobulin-like (BIg-like) domain of ecSODs bind to the cell membrane and transiently open hemichannels, facilitating ROS reductions. RNAi-mediated ecSOD silencing in vivo elevated ROS in host hemocytes, impairing parasitoid larva development. In vitro, the ecSOD-monopolymer needed to be membrane bound to open hemichannels. Furthermore, the hexapep motif in the beta-sandwich of ecSOD49 and ecSOD58, and BIg-like domain in the signal peptides of ecSOD67 were required for cell membrane binding. Hexapep motif and BIg-like domain deletions induced ecSODs loss of adhesion and ROS reduction failure. The hexapep motif and BIg-like domain mediated ecSOD binding via upregulating innexins and stabilizing the opened hemichannels. Our findings reveal a mechanism through which ecSOD reduces ROS, which may aid in developing anti-redox therapy.
Collapse
Affiliation(s)
- Jiang-Hui Meng
- School of Life Sciences, Yunnan University, Kunming, Yunnan 650500, P.R. China
- Yunnan International Joint Laboratory of Virology & Immunology, Kunming, Yunnan 650500, P.R. China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, Yunnan 650500, P.R. China
| | - Yong-Biao Huang
- School of Life Sciences, Yunnan University, Kunming, Yunnan 650500, P.R. China
- Yunnan International Joint Laboratory of Virology & Immunology, Kunming, Yunnan 650500, P.R. China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, Yunnan 650500, P.R. China
| | - Jin Long
- School of Life Sciences, Yunnan University, Kunming, Yunnan 650500, P.R. China
- Yunnan International Joint Laboratory of Virology & Immunology, Kunming, Yunnan 650500, P.R. China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, Yunnan 650500, P.R. China
| | - Qiu-Chen Cai
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, 72076 Tübingen, Germany
| | - Xin Qiao
- School of Life Sciences, Yunnan University, Kunming, Yunnan 650500, P.R. China
- Yunnan International Joint Laboratory of Virology & Immunology, Kunming, Yunnan 650500, P.R. China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, Yunnan 650500, P.R. China
| | - Qiong-Li Zhang
- School of Life Sciences, Yunnan University, Kunming, Yunnan 650500, P.R. China
- Yunnan International Joint Laboratory of Virology & Immunology, Kunming, Yunnan 650500, P.R. China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, Yunnan 650500, P.R. China
| | - Li-Dan Zhang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Xiang Yan
- School of Life Sciences, Yunnan University, Kunming, Yunnan 650500, P.R. China
- Yunnan International Joint Laboratory of Virology & Immunology, Kunming, Yunnan 650500, P.R. China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, Yunnan 650500, P.R. China
| | - Rui Jing
- School of Life Sciences, Yunnan University, Kunming, Yunnan 650500, P.R. China
- Yunnan International Joint Laboratory of Virology & Immunology, Kunming, Yunnan 650500, P.R. China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, Yunnan 650500, P.R. China
| | - Xing-Shan Liu
- School of Life Sciences, Yunnan University, Kunming, Yunnan 650500, P.R. China
- Yunnan International Joint Laboratory of Virology & Immunology, Kunming, Yunnan 650500, P.R. China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, Yunnan 650500, P.R. China
| | - Sai-Jun Zhou
- School of Life Sciences, Yunnan University, Kunming, Yunnan 650500, P.R. China
- Yunnan International Joint Laboratory of Virology & Immunology, Kunming, Yunnan 650500, P.R. China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, Yunnan 650500, P.R. China
| | - Yong-Sheng Yuan
- School of Life Sciences, Yunnan University, Kunming, Yunnan 650500, P.R. China
- Yunnan International Joint Laboratory of Virology & Immunology, Kunming, Yunnan 650500, P.R. China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, Yunnan 650500, P.R. China
| | - Yin-Chen Ma
- School of Life Sciences, Yunnan University, Kunming, Yunnan 650500, P.R. China
- Yunnan International Joint Laboratory of Virology & Immunology, Kunming, Yunnan 650500, P.R. China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, Yunnan 650500, P.R. China
| | - Li-Xiang Zhou
- School of Life Sciences, Yunnan University, Kunming, Yunnan 650500, P.R. China
- Yunnan International Joint Laboratory of Virology & Immunology, Kunming, Yunnan 650500, P.R. China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, Yunnan 650500, P.R. China
| | - Nan-Nan Peng
- School of Life Sciences, Yunnan University, Kunming, Yunnan 650500, P.R. China
- Yunnan International Joint Laboratory of Virology & Immunology, Kunming, Yunnan 650500, P.R. China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, Yunnan 650500, P.R. China
| | - Xing-Cheng Li
- School of Life Sciences, Yunnan University, Kunming, Yunnan 650500, P.R. China
- Yunnan International Joint Laboratory of Virology & Immunology, Kunming, Yunnan 650500, P.R. China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, Yunnan 650500, P.R. China
| | - Cheng-Hui Cai
- School of Life Sciences, Yunnan University, Kunming, Yunnan 650500, P.R. China
- Yunnan International Joint Laboratory of Virology & Immunology, Kunming, Yunnan 650500, P.R. China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, Yunnan 650500, P.R. China
| | - Hong-Mei Tang
- School of Life Sciences, Yunnan University, Kunming, Yunnan 650500, P.R. China
- Yunnan International Joint Laboratory of Virology & Immunology, Kunming, Yunnan 650500, P.R. China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, Yunnan 650500, P.R. China
| | - André F. Martins
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, 72076 Tübingen, Germany
| | - Jean X. Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Kai-Jun Luo
- School of Life Sciences, Yunnan University, Kunming, Yunnan 650500, P.R. China
- Yunnan International Joint Laboratory of Virology & Immunology, Kunming, Yunnan 650500, P.R. China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
3
|
Petsakou A, Liu Y, Liu Y, Comjean A, Hu Y, Perrimon N. Epithelial Ca 2+ waves triggered by enteric neurons heal the gut. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.14.553227. [PMID: 37645990 PMCID: PMC10461974 DOI: 10.1101/2023.08.14.553227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
A fundamental and unresolved question in regenerative biology is how tissues return to homeostasis after injury. Answering this question is essential for understanding the etiology of chronic disorders such as inflammatory bowel diseases and cancer. We used the Drosophila midgut to investigate this question and discovered that during regeneration a subpopulation of cholinergic enteric neurons triggers Ca2+ currents among enterocytes to promote return of the epithelium to homeostasis. Specifically, we found that down-regulation of the cholinergic enzyme Acetylcholinesterase in the epithelium enables acetylcholine from defined enteric neurons, referred as ARCENs, to activate nicotinic receptors in enterocytes found near ARCEN-innervations. This activation triggers high Ca2+ influx that spreads in the epithelium through Inx2/Inx7 gap junctions promoting enterocyte maturation followed by reduction of proliferation and inflammation. Disrupting this process causes chronic injury consisting of ion imbalance, Yki activation and increase of inflammatory cytokines together with hyperplasia, reminiscent of inflammatory bowel diseases. Altogether, we found that during gut regeneration the conserved cholinergic pathway facilitates epithelial Ca2+ waves that heal the intestinal epithelium. Our findings demonstrate nerve- and bioelectric-dependent intestinal regeneration which advance the current understanding of how a tissue returns to its homeostatic state after injury and could ultimately help existing therapeutics.
Collapse
Affiliation(s)
| | - Yifang Liu
- Department of Genetics, Harvard Medical School, Boston, USA
| | - Ying Liu
- Department of Genetics, Harvard Medical School, Boston, USA
| | - Aram Comjean
- Department of Genetics, Harvard Medical School, Boston, USA
| | - Yanhui Hu
- Department of Genetics, Harvard Medical School, Boston, USA
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, USA
- Howard Hughes Medical Institute, Boston, USA
| |
Collapse
|
4
|
Güiza J, Solís F, Valenzuela B, Arancibia D, Zamorano P, González J, Saavedra J, Neely A, Salgado M, Martínez AD, Sáez JC, Vega JL. Unnexin is a protein subunit of a large-pore channel expressed by unicellular organisms. Proc Natl Acad Sci U S A 2023; 120:e2307898120. [PMID: 37487087 PMCID: PMC10400985 DOI: 10.1073/pnas.2307898120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 06/23/2023] [Indexed: 07/26/2023] Open
Abstract
Cells of vertebrate and invertebrate organisms express proteins specialized in membrane channel-based cell-cell communication that are absent in unicellular organisms. We recently described the prediction of some members of the large-pore channel family in kinetoplastids, consisting of proteins called unnexins, which share several structural features with innexin and pannexin proteins. Here, we demonstrated that the unnexin1 protein (Unx1) is delivered to the cell membrane, displaying a topology consisting of four transmembrane domains with C and N termini on the cytoplasmic side and form large-pore channels that are permeable to small molecules. Low extracellular Ca2+/Mg2+ levels or extracellular alkalinization, but not mechanical stretching, increases channel activity. The Unx1 channel mediates the influx of Ca2+ and does not form intercellular dye coupling between HeLa Unx1 transfected cells. Unx1 channel function was further evidenced by its ability to mediate ionic currents when expressed in Xenopus oocytes. Downregulation of Unx1 mRNA with morpholine contains Trypanosoma cruzi invasion. Phylogenetic analysis revealed the presence of Unx1 homologs in other protozoan parasites, suggesting a conserved function for these channel parasites in other protists. Our data demonstrate that Unx1 forms large-pore membrane channels, which may serve as a diffusional pathway for ions and small molecules that are likely to be metabolic substrates or waste products, and signaling autocrine and paracrine molecules that could be involved in cell invasion. As morpholinos-induced downregulation of Unx1 reduces the infectivity of trypomastigotes, the Unx1 channels might be an attractive target for developing trypanocide drugs.
Collapse
Affiliation(s)
- Juan Güiza
- Laboratory of Gap Junction Proteins and Parasitic Diseases (GaPaL), Instituto Antofagasta, Universidad de Antofagasta, Antofagasta1240000, Chile
| | - Francisco Solís
- Laboratorio de Microorganismos Extremófilos, Instituto Antofagasta, Universidad de Antofagasta, Antofagasta1240000, Chile
| | - Bernardita Valenzuela
- Laboratorio de Microorganismos Extremófilos, Instituto Antofagasta, Universidad de Antofagasta, Antofagasta1240000, Chile
| | - Duxan Arancibia
- Laboratorio de Microorganismos Extremófilos, Instituto Antofagasta, Universidad de Antofagasta, Antofagasta1240000, Chile
| | - Pedro Zamorano
- Laboratorio de Microorganismos Extremófilos, Instituto Antofagasta, Universidad de Antofagasta, Antofagasta1240000, Chile
| | - Jorge González
- Departamento de Tecnología Médica, Unidad de Parasitología Molecular, Facultad Ciencias de la Salud, Universidad de Antofagasta, Antofagasta1240000, Chile
| | - Jonathan Saavedra
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2381850, Chile
| | - Alan Neely
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2381850, Chile
| | - Magdiel Salgado
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2381850, Chile
| | - Agustín D. Martínez
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2381850, Chile
| | - Juan C. Sáez
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2381850, Chile
| | - José L. Vega
- Laboratory of Gap Junction Proteins and Parasitic Diseases (GaPaL), Instituto Antofagasta, Universidad de Antofagasta, Antofagasta1240000, Chile
- Departamento Biomédico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta1240000, Chile
| |
Collapse
|
5
|
Caña-Bozada V, Morales-Serna FN, Fajer-Ávila EJ, Llera-Herrera R. De novo transcriptome assembly and identification of G-Protein-Coupled-Receptors (GPCRs) in two species of monogenean parasites of fish. Parasite 2022; 29:51. [PMID: 36350193 PMCID: PMC9645230 DOI: 10.1051/parasite/2022052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 10/13/2022] [Indexed: 11/11/2022] Open
Abstract
Genomic resources for Platyhelminthes of the class Monogenea are scarce, despite the diversity of these parasites, some species of which are highly pathogenic to their fish hosts. This work aimed to generate de novo-assembled transcriptomes of two monogenean species, Scutogyrus longicornis (Dactylogyridae) and Rhabdosynochus viridisi (Diplectanidae), providing a protocol for cDNA library preparation with low input samples used in single cell transcriptomics. This allowed us to work with sub-microgram amounts of total RNA with success. These transcriptomes consist of 25,696 and 47,187 putative proteins, respectively, which were further annotated according to the Swiss-Prot, Pfam, GO, KEGG, and COG databases. The completeness values of these transcriptomes evaluated with BUSCO against Metazoa databases were 54.1% and 73%, respectively, which is in the range of other monogenean species. Among the annotations, a large number of terms related to G-protein-coupled receptors (GPCRs) were found. We identified 109 GPCR-like sequences in R. viridisi, and 102 in S. longicornis, including family members specific for Platyhelminthes. Rhodopsin was the largest family according to GRAFS classification. Two putative melatonin receptors found in S. longicornis represent the first record of this group of proteins in parasitic Platyhelminthes. Forty GPCRs of R. viridisi and 32 of S. longicornis that were absent in Vertebrata might be potential drug targets. The present study provides the first publicly available transcriptomes for monogeneans of the subclass Monopisthocotylea, which can serve as useful genomic datasets for functional genomic research of this important group of parasites.
Collapse
Affiliation(s)
- Víctor Caña-Bozada
- Centro de Investigación en Alimentación y Desarrollo, A.C. Unidad Mazatlán en Acuicultura y Manejo Ambiental Mazatlán Sinaloa 82112 Mexico
| | - F. Neptalí Morales-Serna
- Instituto de Ciencias del Mar y Limnología, Unidad Académica Mazatlán, Universidad Nacional Autónoma de México Mazatlán Sinaloa 82040 Mexico
| | - Emma J. Fajer-Ávila
- Centro de Investigación en Alimentación y Desarrollo, A.C. Unidad Mazatlán en Acuicultura y Manejo Ambiental Mazatlán Sinaloa 82112 Mexico
| | - Raúl Llera-Herrera
- Instituto de Ciencias del Mar y Limnología, Unidad Académica Mazatlán, Universidad Nacional Autónoma de México Mazatlán Sinaloa 82040 Mexico
| |
Collapse
|
6
|
Belardin LB, Brochu K, Légaré C, Battistone MA, Breton S. Purinergic signaling in the male reproductive tract. Front Endocrinol (Lausanne) 2022; 13:1049511. [PMID: 36419764 PMCID: PMC9676935 DOI: 10.3389/fendo.2022.1049511] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 10/21/2022] [Indexed: 11/09/2022] Open
Abstract
Purinergic receptors are ubiquitously expressed throughout the body and they participate in the autocrine and paracrine regulation of cell function during normal physiological and pathophysiological conditions. Extracellular nucleotides activate several types of plasma membrane purinergic receptors that form three distinct families: P1 receptors are activated by adenosine, P2X receptors are activated by ATP, and P2Y receptors are activated by nucleotides including ATP, ADP, UTP, UDP, and UDP-glucose. These specific pharmacological fingerprints and the distinct intracellular signaling pathways they trigger govern a large variety of cellular responses in an organ-specific manner. As such, purinergic signaling regulates several physiological cell functions, including cell proliferation, differentiation and death, smooth muscle contraction, vasodilatation, and transepithelial transport of water, solute, and protons, as well as pathological pathways such as inflammation. While purinergic signaling was first discovered more than 90 years ago, we are just starting to understand how deleterious signals mediated through purinergic receptors may be involved in male infertility. A large fraction of male infertility remains unexplained illustrating our poor understanding of male reproductive health. Purinergic signaling plays a variety of physiological and pathophysiological roles in the male reproductive system, but our knowledge in this context remains limited. This review focuses on the distribution of purinergic receptors in the testis, epididymis, and vas deferens, and their role in the establishment and maintenance of male fertility.
Collapse
Affiliation(s)
- Larissa Berloffa Belardin
- Centre Hospitalier Universitaire de Québec - Research Centre and Department of Obstetrics, Gynecology and Reproduction, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Kéliane Brochu
- Centre Hospitalier Universitaire de Québec - Research Centre and Department of Obstetrics, Gynecology and Reproduction, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Christine Légaré
- Centre Hospitalier Universitaire de Québec - Research Centre and Department of Obstetrics, Gynecology and Reproduction, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Maria Agustina Battistone
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Sylvie Breton
- Centre Hospitalier Universitaire de Québec - Research Centre and Department of Obstetrics, Gynecology and Reproduction, Faculty of Medicine, Université Laval, Québec, QC, Canada
| |
Collapse
|
7
|
Wang J, Chen X, Hou X, Wang J, Yue W, Huang S, Xu G, Yan J, Lu G, Hofreiter M, Li C, Wang C. "Omics" data unveil early molecular response underlying limb regeneration in the Chinese mitten crab, Eriocheir sinensis. SCIENCE ADVANCES 2022; 8:eabl4642. [PMID: 36112682 PMCID: PMC9481118 DOI: 10.1126/sciadv.abl4642] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/01/2022] [Indexed: 05/22/2023]
Abstract
Limb regeneration is a fascinating and medically interesting trait that has been well preserved in arthropod lineages, particularly in crustaceans. However, the molecular mechanisms underlying arthropod limb regeneration remain largely elusive. The Chinese mitten crab Eriocheir sinensis shows strong regenerative capacity, a trait that has likely allowed it to become a worldwide invasive species. Here, we report a chromosome-level genome of E. sinensis as well as large-scale transcriptome data during the limb regeneration process. Our results reveal that arthropod-specific genes involved in signal transduction, immune response, histone methylation, and cuticle development all play fundamental roles during the regeneration process. Particularly, Innexin2-mediated signal transduction likely facilitates the early stage of the regeneration process, while an effective crustacean-specific prophenoloxidase system (ProPo-AS) plays crucial roles in the initial immune response. Collectively, our findings uncover novel genetic pathways pertaining to arthropod limb regeneration and provide valuable resources for studies on regeneration from a comparative perspective.
Collapse
Affiliation(s)
- Jun Wang
- Key Laboratory of Freshwater Aquatic Genetic Resources certified by the Ministry of Agriculture and Rural Affairs/National Demonstration Center for Experimental Fisheries Science Education/Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| | - Xiaowen Chen
- Key Laboratory of Freshwater Aquatic Genetic Resources certified by the Ministry of Agriculture and Rural Affairs/National Demonstration Center for Experimental Fisheries Science Education/Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| | - Xin Hou
- Key Laboratory of Freshwater Aquatic Genetic Resources certified by the Ministry of Agriculture and Rural Affairs/National Demonstration Center for Experimental Fisheries Science Education/Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| | - Jingan Wang
- Key Laboratory of Freshwater Aquatic Genetic Resources certified by the Ministry of Agriculture and Rural Affairs/National Demonstration Center for Experimental Fisheries Science Education/Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| | - Wucheng Yue
- Key Laboratory of Freshwater Aquatic Genetic Resources certified by the Ministry of Agriculture and Rural Affairs/National Demonstration Center for Experimental Fisheries Science Education/Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| | - Shu Huang
- Key Laboratory of Freshwater Aquatic Genetic Resources certified by the Ministry of Agriculture and Rural Affairs/National Demonstration Center for Experimental Fisheries Science Education/Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| | - Gangchun Xu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization certified by the Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Jizhou Yan
- Key Laboratory of Freshwater Aquatic Genetic Resources certified by the Ministry of Agriculture and Rural Affairs/National Demonstration Center for Experimental Fisheries Science Education/Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| | - Guoqing Lu
- Department of Biology, University of Nebraska at Omaha, Omaha, NE 68182, USA
| | - Michael Hofreiter
- Evolutionary Adaptive Genomics, Institute of Biochemistry and Biology, Faculty of Science, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476 Potsdam, Germany
- Corresponding author. Email (M.H.); (C.L.); (C.W.)
| | - Chenhong Li
- Key Laboratory of Freshwater Aquatic Genetic Resources certified by the Ministry of Agriculture and Rural Affairs/National Demonstration Center for Experimental Fisheries Science Education/Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
- Corresponding author. Email (M.H.); (C.L.); (C.W.)
| | - Chenghui Wang
- Key Laboratory of Freshwater Aquatic Genetic Resources certified by the Ministry of Agriculture and Rural Affairs/National Demonstration Center for Experimental Fisheries Science Education/Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
- Corresponding author. Email (M.H.); (C.L.); (C.W.)
| |
Collapse
|
8
|
Ugur D, Gungul TB, Yucel S, Ozcivici E, Yalcin-Ozuysal O, Mese G. Connexin 32 overexpression increases proliferation, reduces gap junctional intercellular communication, motility and epithelial-to-mesenchymal transition in Hs578T breast cancer cells. J Cell Commun Signal 2022; 16:361-376. [PMID: 35781670 DOI: 10.1007/s12079-021-00665-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 12/14/2021] [Indexed: 11/29/2022] Open
Abstract
Connexins (Cx) are primary components of gap junctions that selectively allow molecules to be exchanged between adjacent cells, regulating multiple cellular functions. Along with their channel forming functions, connexins play a variety of roles in different stages of tumorigenesis and their roles in tumor initiation and progression is isoform- and tissue-specific. While Cx26 and Cx43 were downregulated during breast tumorigenesis, Cx32 was accumulated in the cytoplasm of the cells in lymph node metastasis of breast cancers and Cx32 was further upregulated in metastasis. Cx32's effect on cell proliferation, gap junctional communication, hemichannel activity, cellular motility and epithelial-to-mesenchymal transition (EMT) were investigated by overexpressing Cx32 in Hs578T and MCF7 breast cancer cells. Additionally, the expression and localization of Cx26 and Cx43 upon Cx32 overexpression were examined by Western blot and immunostaining experiments, respectively. We observed that MCF7 cells had endogenous Cx32 while Hs578T cells did not and when Cx32 was overexpressed in these cells, it caused a significant increase in the percentages of Hs578T cells at the S phase in addition to increasing their proliferation. Further, while Cx32 overexpression did not induce hemichannel activity in either cell, it decreased gap junctional communication between Hs578T cells. Additionally, Cx32 was mainly observed in the cytoplasm in both cells, where it did not form gap junction plaques but Cx32 overexpression reduced Cx43 levels without affecting Cx26. Moreover, migration and invasion potentials of Hs578T and migration in MCF7 were reduced upon Cx32 overexpression. Finally, the protein level of mesenchymal marker N-cadherin decreased while epithelial marker ZO-1 and E-cadherin increased in Hs578T cells. We observed that Cx32 overexpression altered cell proliferation, communication, migration and EMT in Hs578T, suggesting a tumor suppressor role in these cells while it had minor effects on MCF7 cells.
Collapse
Affiliation(s)
- Deniz Ugur
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir, 35430, Turkey.,Department of Molecular Biology and Genetics, Avrasya University, Trabzon, Turkey
| | - Taha Bugra Gungul
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir, 35430, Turkey
| | - Simge Yucel
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir, 35430, Turkey
| | - Engin Ozcivici
- Department of Bioengineering, Izmir Institute of Technology, Urla, Izmir, Turkey
| | - Ozden Yalcin-Ozuysal
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir, 35430, Turkey
| | - Gulistan Mese
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Urla, Izmir, 35430, Turkey.
| |
Collapse
|
9
|
Welzel G, Schuster S. Connexins evolved after early chordates lost innexin diversity. eLife 2022; 11:74422. [PMID: 35042580 PMCID: PMC8769644 DOI: 10.7554/elife.74422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/05/2022] [Indexed: 11/26/2022] Open
Abstract
Gap junction channels are formed by two unrelated protein families. Non-chordates use the primordial innexins, while chordates use connexins that superseded the gap junction function of innexins. Chordates retained innexin-homologs, but N-glycosylation prevents them from forming gap junctions. It is puzzling why chordates seem to exclusively use the new gap junction protein and why no chordates should exist that use non-glycosylated innexins to form gap junctions. Here, we identified glycosylation sites of 2388 innexins from 174 non-chordate and 276 chordate species. Among all chordates, we found not a single innexin without glycosylation sites. Surprisingly, the glycosylation motif is also widespread among non-chordate innexins indicating that glycosylated innexins are not a novelty of chordates. In addition, we discovered a loss of innexin diversity during early chordate evolution. Most importantly, lancelets, which lack connexins, exclusively possess only one highly conserved innexin with one glycosylation site. A bottleneck effect might thus explain why connexins have become the only protein used to form chordate gap junctions.
Collapse
Affiliation(s)
- Georg Welzel
- Department of Animal Physiology, University of Bayreuth
| | | |
Collapse
|
10
|
Zhao J, Zhang W, Wu T, Wang H, Mao J, Liu J, Zhou Z, Lin X, Yan H, Wang Q. Efferocytosis in the Central Nervous System. Front Cell Dev Biol 2021; 9:773344. [PMID: 34926460 PMCID: PMC8678611 DOI: 10.3389/fcell.2021.773344] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/25/2021] [Indexed: 11/23/2022] Open
Abstract
The effective clearance of apoptotic cells is essential for maintaining central nervous system (CNS) homeostasis and restoring homeostasis after injury. In most cases of physiological apoptotic cell death, efferocytosis prevents inflammation and other pathological conditions. When apoptotic cells are not effectively cleared, destruction of the integrity of the apoptotic cell membrane integrity, leakage of intracellular contents, and secondary necrosis may occur. Efferocytosis is the mechanism by which efferocytes quickly remove apoptotic cells from tissues before they undergo secondary necrosis. Cells with efferocytosis functions, mainly microglia, help to eliminate apoptotic cells from the CNS. Here, we discuss the impacts of efferocytosis on homeostasis, the mechanism of efferocytosis, the associations of efferocytosis failure and CNS diseases, and the current clinical applications of efferocytosis. We also identify efferocytosis as a novel potential target for exploring the causes and treatments of CNS diseases.
Collapse
Affiliation(s)
- Jiayi Zhao
- Department of Anesthesia, Zhejiang Hospital, Hangzhou, China
| | - Weiqi Zhang
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Tingting Wu
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Hongyi Wang
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jialiang Mao
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jian Liu
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ziheng Zhou
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xianfeng Lin
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huige Yan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingqing Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
11
|
Syrjanen J, Michalski K, Kawate T, Furukawa H. On the molecular nature of large-pore channels. J Mol Biol 2021; 433:166994. [PMID: 33865869 PMCID: PMC8409005 DOI: 10.1016/j.jmb.2021.166994] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 04/08/2021] [Accepted: 04/08/2021] [Indexed: 12/25/2022]
Abstract
Membrane transport is a fundamental means to control basic cellular processes such as apoptosis, inflammation, and neurodegeneration and is mediated by a number of transporters, pumps, and channels. Accumulating evidence over the last half century has shown that a type of so-called "large-pore channel" exists in various tissues and organs in gap-junctional and non-gap-junctional forms in order to flow not only ions but also metabolites such as ATP. They are formed by a number of protein families with little or no evolutionary linkages including connexin, innexin, pannexin, leucine-rich repeat-containing 8 (LRRC8), and calcium homeostasis modulator (CALHM). This review summarizes the history and concept of large-pore channels starting from connexin gap junction channels to the more recent developments in innexin, pannexin, LRRC8, and CALHM. We describe structural and functional features of large-pore channels that are crucial for their diverse functions on the basis of available structures.
Collapse
Affiliation(s)
- Johanna Syrjanen
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Kevin Michalski
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Toshimitsu Kawate
- Department of Molecular Medicine, Fields of Biochemistry, Molecular, and Cell Biology (BMCB), and Biophysics, Cornell University, Ithaca, NY 14853, USA
| | - Hiro Furukawa
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
12
|
Cárcaba L, García-Piqueras J, García-Mesa Y, Cobo R, García-Suárez O, Feito J, Vega JA. Human digital merkel cells display pannexin1 immunoreactivity. Ann Anat 2021; 239:151813. [PMID: 34384856 DOI: 10.1016/j.aanat.2021.151813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 10/20/2022]
Abstract
Pannexins are channel proteins displaying functional similarities to gap junctions in vertebrates and are regarded as transmembrane ATP-releasing channels. A member of this family, denominate pannexin1, has been detected in the epidermis and cutaneous adnexal structures. Here we used immunohistochemistry to investigate whether human digital Merkel cells express this protein since ATP is postulated as a neurotransmitter in the Merkel cell-axon complexes low-threshold mecahoreceptors. Pannexin1 immunoreactivity was found in cytokeratine 20-, chromogranin A- and synaptophysin-positive cells placed at the basal layer of the epidermis. Cell displaying pannexin1 immunoreactivities were thus identified as Merkel cells and showed close contact with nerve profiles. Light pannexin1 immunoreactivity in dermal blood vessels was also verified. Present results demonstrate for the first time the expression of pannexin1 in human digital Merkel cells supporting the idea that ATP can be involved directly or indirectly in the mechanotransductional process at Merkel-axon complexes.
Collapse
Affiliation(s)
- Lucia Cárcaba
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de Oviedo, Spain
| | - Jorge García-Piqueras
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de Oviedo, Spain
| | - Yolanda García-Mesa
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de Oviedo, Spain
| | - Ramón Cobo
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de Oviedo, Spain
| | - Olivia García-Suárez
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de Oviedo, Spain
| | - Jorge Feito
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de Oviedo, Spain; Servicio de Anatomía Patológica, Complejo Hospitalario Universitario de Salamanca, Salamanca, Spain; Departamento de Anatomía e Histología Humanas, Universidad de Salamanca, Salamanca, Spain
| | - José A Vega
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de Oviedo, Spain; Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago de Chile, Chile.
| |
Collapse
|
13
|
Sayegh MN, Cooney KA, Han WM, Wang L, Strobel F, Hansen LM, García AJ, Levit RD. A Hydrogel Strategy to Augment Tissue Adenosine to Improve Hindlimb Perfusion. Arterioscler Thromb Vasc Biol 2021; 41:e314-e324. [PMID: 33882686 PMCID: PMC8159890 DOI: 10.1161/atvbaha.120.315428] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Michael N. Sayegh
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA
| | - Kimberly A. Cooney
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Woojin M. Han
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Lanfang Wang
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | | | - Laura M. Hansen
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Andrés J. García
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA
| | - Rebecca D. Levit
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
14
|
Carbenoxolone has the potential to ameliorate acute incision pain in rats. Mol Med Rep 2021; 24:520. [PMID: 34013377 PMCID: PMC8160483 DOI: 10.3892/mmr.2021.12159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 01/07/2021] [Indexed: 12/23/2022] Open
Abstract
Carbenoxolone (CBX) is primarily used to relieve various types of neuropathic and inflammatory pain. However, little is known concerning the role of CBX in acute pain and its functional mechanisms therein and this was investigated in the present study. Rats underwent toe incision and behavioral tests were performed to assess mechanical hypersensitivity. The expression levels of pannexin 1 (Px1) and connexin 43 (Cx43) were detected using western blot analysis 2, 4, 6 or 24 h after toe incision, and the expression of TNF-α, IL-1β and P substance (SP) was determined by ELISA; Px1 and Cx43 expression was also examined by immunofluorescence staining. At 2, 6 and 12 h post-toe incision, the postoperative pain threshold was significantly reduced, which was subsequently recovered at 2 and 6 h post-surgery following pretreatment with CBX or pannexin 1 mimetic inhibitory peptide. CBX reduced Px1 levels at 4 and 24 h post-incision. However, Cx43 levels were reduced by CBX as little as 2 h post-surgery. Furthermore, CBX not only distinctly decreased the levels of Px1 and Cx43, but also reduced the co-localization of Px1 or Cx43 with glial fibrillary acidic protein, 2 h after incision. It was also observed that the protein levels of inflammatory makers (IL-1β, SP and TNF-α) showed a tendency to decline at 2, 4, 6 and 24 h after incision. Collectively, the expression of Px1 and Cx43 in astrocytes may be involved in pain behaviors diminished by CBX, and CBX potentially reduces acute pain by decreasing Px1 and Cx43 levels. Px1 and Cx43 from spinal astrocytes may serve important roles in the early stages and maintenance of acute pain, while preoperative injection of CBX has the potential to relieve hyperalgesia.
Collapse
|
15
|
Mim C, Perkins G, Dahl G. Structure versus function: Are new conformations of pannexin 1 yet to be resolved? J Gen Physiol 2021; 153:e202012754. [PMID: 33835130 PMCID: PMC8042604 DOI: 10.1085/jgp.202012754] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pannexin 1 (Panx1) plays a decisive role in multiple physiological and pathological settings, including oxygen delivery to tissues, mucociliary clearance in airways, sepsis, neuropathic pain, and epilepsy. It is widely accepted that Panx1 exerts its role in the context of purinergic signaling by providing a transmembrane pathway for ATP. However, under certain conditions, Panx1 can also act as a highly selective membrane channel for chloride ions without ATP permeability. A recent flurry of publications has provided structural information about the Panx1 channel. However, while these structures are consistent with a chloride selective channel, none show a conformation with strong support for the ATP release function of Panx1. In this Viewpoint, we critically assess the existing evidence for the function and structure of the Panx1 channel and conclude that the structure corresponding to the ATP permeation pathway is yet to be determined. We also list a set of additional topics needing attention and propose ways to attain the large-pore, ATP-permeable conformation of the Panx1 channel.
Collapse
Affiliation(s)
- Carsten Mim
- Department of Biomedical Engineering and Health Systems Royal Institute of Technology, Huddinge, Sweden
| | - Guy Perkins
- National Center for Microscopy and Imaging Research, University of California, San Diego School of Medicine, La Jolla, CA
| | - Gerhard Dahl
- Department of Physiology, University of Miami School of Medicine, Miami, FL
| |
Collapse
|
16
|
Imamura H, Sakamoto S, Yoshida T, Matsui Y, Penuela S, Laird DW, Mizukami S, Kikuchi K, Kakizuka A. Single-cell dynamics of pannexin-1-facilitated programmed ATP loss during apoptosis. eLife 2020; 9:61960. [PMID: 33052098 PMCID: PMC7556867 DOI: 10.7554/elife.61960] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 09/25/2020] [Indexed: 12/16/2022] Open
Abstract
ATP is essential for all living cells. However, how dead cells lose ATP has not been well investigated. In this study, we developed new FRET biosensors for dual imaging of intracellular ATP level and caspase-3 activity in single apoptotic cultured human cells. We show that the cytosolic ATP level starts to decrease immediately after the activation of caspase-3, and this process is completed typically within 2 hr. The ATP decrease was facilitated by caspase-dependent cleavage of the plasma membrane channel pannexin-1, indicating that the intracellular decrease of the apoptotic cell is a 'programmed' process. Apoptotic cells deficient of pannexin-1 sustained the ability to produce ATP through glycolysis and to consume ATP, and did not stop wasting glucose much longer period than normal apoptotic cells. Thus, the pannexin-1 plays a role in arresting the metabolic activity of dead apoptotic cells, most likely through facilitating the loss of intracellular ATP.
Collapse
Affiliation(s)
- Hiromi Imamura
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | | | - Tomoki Yoshida
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Yusuke Matsui
- Graduate School of Engineering, Osaka University, Suita, Japan
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Dale W Laird
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Shin Mizukami
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Sendai, Japan
| | - Kazuya Kikuchi
- Graduate School of Engineering, Osaka University, Suita, Japan
| | - Akira Kakizuka
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
17
|
Segura OM, Abdulnoor L, Hua VV, Solano MJ, Macagno ER, Baker MW. Purinergic modulation of neuronal gap junction circuits in the CNS of the leech. J Neurosci Res 2020; 98:1232-1249. [PMID: 32096570 DOI: 10.1002/jnr.24599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/25/2020] [Accepted: 02/05/2020] [Indexed: 11/08/2022]
Abstract
Gap junctions (GJs) are widely distributed in brains across the animal kingdom. To visualize the GJ- coupled networks of two major mechanosensory neurons in the ganglia of medicinal leeches, we injected these cells with the GJ-permeable tracer Neurobiotin. When diffusion time was limited to only 30 min, tracer coupling was highly variable for both cells, suggesting a possible modulation of GJ permeability. In invertebrates the innexins (homologs of vertebrate pannexins) form the GJs. Because extracellular adenosine triphosphate (ATP) modulates pannexin and leech innexin hemichannel permeability and is released by leech glial cells following injury, we tested the effects of bath application of ATP after the injection of Neurobiotin and observed a significant increase in the number of neurons tracer coupled to the sensory neurons. This effect required the elevation of intracellular Ca2+ and could be produced by bath application of caffeine. Conversely, scavenging endogenous extracellular ATP with the ATPase apyrase decreased the number of coupled cells. ATP also increased electrical conductance and tracer permeability between the bilateral Retzius neurons. This modulatory effect of ATP on GJ coupling was blocked by siRNA knockdown of a P1-like adenosine receptor. Finally, exposure of leech ganglia to extracellular ATP induced a characteristic low frequency (<0.3 Hz) rhythmic bursting activity that was roughly synchronous among multiple neurons, a behavior that was significantly attenuated by the GJ blocker octanol. These findings highlight the mediation by ATP of a robust physiological mechanism for modifying neuronal circuits by rapidly recruiting neurons into active networks and entraining synchronized bursting activity.
Collapse
Affiliation(s)
- Oliva Mota Segura
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Lina Abdulnoor
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Vinh-Vincent Hua
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Martha J Solano
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Eduardo R Macagno
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Michael W Baker
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA.,Department of Psychology, Mount Saint Vincent University, Halifax, Nova Scotia, Canada
| |
Collapse
|
18
|
Role of an Aromatic-Aromatic Interaction in the Assembly and Trafficking of the Zebrafish Panx1a Membrane Channel. Biomolecules 2020; 10:biom10020272. [PMID: 32053881 PMCID: PMC7072349 DOI: 10.3390/biom10020272] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/29/2020] [Accepted: 02/05/2020] [Indexed: 12/28/2022] Open
Abstract
Pannexin 1 (Panx1) is a ubiquitously expressed hexameric integral membrane protein known to function as an adenosine triphosphate (ATP) release channel. Panx1 proteins exist in unglycosylated core form (Gly0). They undergo critical post-translational modifications forming the high mannose glycosylation state (Gly1) in the endoplasmic reticulum (ER) and the complex glycosylation state (Gly2) in the Golgi apparatus. The regulation of transition from the ER to the cell membrane is not fully understood. Using site-specific mutagenesis, dye uptake assays, and interaction testing, we identified two conserved aromatic residues, Trp123 and Tyr205, in the transmembrane domains 2 and 3 of the zebrafish panx1a protein. Results suggest that both residues primarily govern the assembly of panx1a subunits into channels, with mutant proteins failing to interact. The results provide insight into a mechanism enabling regulation of Panx1 oligomerization, glycosylation, and trafficking.
Collapse
|
19
|
Aguilar-Perez A, Pacheco-Costa R, Atkinson EG, Deosthale P, Davis HM, Essex AL, Dilley JE, Gomez L, Rupert JE, Zimmers TA, Thompson RJ, Allen MR, Plotkin LI. Age- and sex-dependent role of osteocytic pannexin1 on bone and muscle mass and strength. Sci Rep 2019; 9:13903. [PMID: 31554905 PMCID: PMC6761284 DOI: 10.1038/s41598-019-50444-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 09/11/2019] [Indexed: 12/25/2022] Open
Abstract
Pannexins (Panxs), glycoproteins that oligomerize to form hemichannels on the cell membrane, are topologically similar to connexins, but do not form cell-to-cell gap junction channels. There are 3 members of the family, 1-3, with Panx1 being the most abundant. All Panxs are expressed in bone, but their role in bone cell biology is not completely understood. We now report that osteocytic Panx1 deletion (Panx1Δot) alters bone mass and strength in female mice. Bone mineral density after reaching skeletal maturity is higher in female Panx1Δot mice than in control Panx1fl/fl mice. Further, osteocytic Panx1 deletion partially prevented aging effects on cortical bone structure and mechanical properties. Young 4-month-old female Panx1Δot mice exhibited increased lean body mass, even though pannexin levels in skeletal muscle were not affected; whereas no difference in lean body mass was detected in male mice. Furthermore, female Panx1-deficient mice exhibited increased muscle mass without changes in strength, whereas Panx1Δot males showed unchanged muscle mass and decreased in vivo maximum plantarflexion torque, indicating reduced muscle strength. Our results suggest that osteocytic Panx1 deletion increases bone mass in young and old female mice and muscle mass in young female mice, but has deleterious effects on muscle strength only in males.
Collapse
Affiliation(s)
- Alexandra Aguilar-Perez
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Rafael Pacheco-Costa
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Emily G Atkinson
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Padmini Deosthale
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Hannah M Davis
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Alyson L Essex
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Julian E Dilley
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Leland Gomez
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Joseph E Rupert
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Teresa A Zimmers
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana Center for Musculoskeletal Health, Indianapolis, IN, 46202, USA
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Indiana University Simon Cancer Center, Indianapolis, IN, 46202, USA
| | - Roger J Thompson
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
| | - Matthew R Allen
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Roudebush Veterans Administration Medical Center, Indianapolis, IN, 46202, USA
| | - Lilian I Plotkin
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Roudebush Veterans Administration Medical Center, Indianapolis, IN, 46202, USA.
- Indiana Center for Musculoskeletal Health, Indianapolis, IN, 46202, USA.
| |
Collapse
|
20
|
Flynn AF, Joyce MG, Taylor RT, Bennuru S, Lindrose AR, Sterling SL, Morris CP, Nutman TB, Mitre E. Intestinal UDP-glucuronosyltransferase as a potential target for the treatment and prevention of lymphatic filariasis. PLoS Negl Trop Dis 2019; 13:e0007687. [PMID: 31513587 PMCID: PMC6742224 DOI: 10.1371/journal.pntd.0007687] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 08/05/2019] [Indexed: 11/19/2022] Open
Abstract
Lymphatic filariasis (LF), a morbid disease caused by the tissue-invasive nematodes Wuchereria bancrofti, Brugia malayi, and Brugia timori, affects millions of people worldwide. Global eradication efforts have significantly reduced worldwide prevalence, but complete elimination has been hampered by limitations of current anti-filarial drugs and the lack of a vaccine. The goal of this study was to evaluate B. malayi intestinal UDP-glucuronosyltransferase (Bm-UGT) as a potential therapeutic target. To evaluate whether Bm-UGT is essential for adult filarial worms, we inhibited its expression using siRNA. This resulted in a 75% knockdown of Bm-ugt mRNA for 6 days and almost complete suppression of detectable Bm-UGT by immunoblot. Reduction in Bm-UGT expression resulted in decreased worm motility for 6 days, 70% reduction in microfilaria release from adult worms, and significant reduction in adult worm metabolism as detected by MTT assays. Because prior allergic-sensitization to a filarial antigen would be a contraindication for its use as a vaccine candidate, we tested plasma from infected and endemic normal populations for Bm-UGT-specific IgE using a luciferase immunoprecipitation assay. All samples (n = 35) tested negative. We then tested two commercially available medicines known to be broad inhibitors of UGTs, sulfinpyrazone and probenecid, for in vitro activity against B. malayi. There were marked macrofilaricidal effects at concentrations achievable in humans and very little effect on microfilariae. In addition, we observed that probenecid and sulfinpyrazone exhibit a synergistic macrofilaricidal effect when used in combination with albendazole. The results of this study demonstrate that Bm-UGT is an essential protein for adult worm survival. Lack of prior IgE sensitization in infected and endemic populations suggest it may be a feasible vaccine candidate. The finding that sulfinpyrazone and probenecid have in vitro effects against adult B. malayi worms suggests that these medications have promise as potential macrofilaricides in humans.
Collapse
Affiliation(s)
- Alexander F. Flynn
- Department of Microbiology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - M. Gordon Joyce
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| | - Rebekah T. Taylor
- Department of Biology, Frostburg State University, Frostburg, Maryland, United States of America
| | - Sasisekhar Bennuru
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alyssa R. Lindrose
- Department of Microbiology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Spencer L. Sterling
- Department of Microbiology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - C. Paul Morris
- Department of Pathology, Johns Hopkins Hospital, Baltimore Maryland, United States of America
| | - Thomas B. Nutman
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Edward Mitre
- Department of Microbiology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| |
Collapse
|
21
|
Beckmann A, Hainz N, Tschernig T, Meier C. Facets of Communication: Gap Junction Ultrastructure and Function in Cancer Stem Cells and Tumor Cells. Cancers (Basel) 2019; 11:cancers11030288. [PMID: 30823688 PMCID: PMC6468480 DOI: 10.3390/cancers11030288] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 02/22/2019] [Accepted: 02/25/2019] [Indexed: 12/28/2022] Open
Abstract
Gap junction proteins are expressed in cancer stem cells and non-stem cancer cells of many tumors. As the morphology and assembly of gap junction channels are crucial for their function in intercellular communication, one focus of our review is to outline the data on gap junction plaque morphology available for cancer cells. Electron microscopic studies and freeze-fracture analyses on gap junction ultrastructure in cancer are summarized. As the presence of gap junctions is relevant in solid tumors, we exemplarily outline their role in glioblastomas and in breast cancer. These were also shown to contain cancer stem cells, which are an essential cause of tumor onset and of tumor transmission into metastases. For these processes, gap junctional communication was shown to be important and thus we summarize, how the expression of gap junction proteins and the resulting communication between cancer stem cells and their surrounding cells contributes to the dissemination of cancer stem cells via blood or lymphatic vessels. Based on their importance for tumors and metastases, future cancer-specific therapies are expected to address gap junction proteins. In turn, gap junctions also seem to contribute to the unattainability of cancer stem cells by certain treatments and might thus contribute to therapeutic resistance.
Collapse
Affiliation(s)
- Anja Beckmann
- Department of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Germany.
| | - Nadine Hainz
- Department of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Germany.
| | - Thomas Tschernig
- Department of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Germany.
| | - Carola Meier
- Department of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Germany.
| |
Collapse
|
22
|
Boesmans W, Hao MM, Fung C, Li Z, Van den Haute C, Tack J, Pachnis V, Vanden Berghe P. Structurally defined signaling in neuro-glia units in the enteric nervous system. Glia 2019; 67:1167-1178. [PMID: 30730592 PMCID: PMC6593736 DOI: 10.1002/glia.23596] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 01/11/2019] [Accepted: 01/11/2019] [Indexed: 12/21/2022]
Abstract
Coordination of gastrointestinal function relies on joint efforts of enteric neurons and glia, whose crosstalk is vital for the integration of their activity. To investigate the signaling mechanisms and to delineate the spatial aspects of enteric neuron-to-glia communication within enteric ganglia we developed a method to stimulate single enteric neurons while monitoring the activity of neighboring enteric glial cells. We combined cytosolic calcium uncaging of individual enteric neurons with calcium imaging of enteric glial cells expressing a genetically encoded calcium indicator and demonstrate that enteric neurons signal to enteric glial cells through pannexins using paracrine purinergic pathways. Sparse labeling of enteric neurons and high-resolution analysis of the structural relation between neuronal cell bodies, varicose release sites and enteric glia uncovered that this form of neuron-to-glia communication is contained between the cell body of an enteric neuron and its surrounding enteric glial cells. Our results reveal the spatial and functional foundation of neuro-glia units as an operational cellular assembly in the enteric nervous system.
Collapse
Affiliation(s)
- Werend Boesmans
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium.,Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands.,Biomedical Research Institute (BIOMED), Hasselt University, Hasselt, Belgium
| | - Marlene M Hao
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium.,Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Australia
| | - Candice Fung
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - Zhiling Li
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - Chris Van den Haute
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, University of Leuven, Leuven, Belgium.,Leuven Viral Vector Core, University of Leuven, Leuven, Belgium
| | - Jan Tack
- Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - Vassilis Pachnis
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| |
Collapse
|
23
|
Linden J, Koch-Nolte F, Dahl G. Purine Release, Metabolism, and Signaling in the Inflammatory Response. Annu Rev Immunol 2019; 37:325-347. [PMID: 30676821 DOI: 10.1146/annurev-immunol-051116-052406] [Citation(s) in RCA: 223] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
ATP, NAD+, and nucleic acids are abundant purines that, in addition to having critical intracellular functions, have evolved extracellular roles as danger signals released in response to cell lysis, apoptosis, degranulation, or membrane pore formation. In general ATP and NAD+ have excitatory and adenosine has anti-inflammatory effects on immune cells. This review focuses on recent advances in our understanding of purine release mechanisms, ectoenzymes that metabolize purines (CD38, CD39, CD73, ENPP1, and ENPP2/autotaxin), and signaling by key P2 purinergic receptors (P2X7, P2Y2, and P2Y12). In addition to metabolizing ATP or NAD+, some purinergic ectoenzymes metabolize other inflammatory modulators, notably lysophosphatidic acid and cyclic GMP-AMP (cGAMP). Also discussed are extracellular signaling effects of NAD+ mediated by ADP-ribosylation, and epigenetic effects of intracellular adenosine mediated by modification of S-adenosylmethionine-dependent DNA methylation.
Collapse
Affiliation(s)
- Joel Linden
- Division of Developmental Immunology, La Jolla Institute for Immunology, La Jolla, California 92037, USA; .,Department of Pharmacology, University of California, San Diego, La Jolla, California 92093, USA
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany;
| | - Gerhard Dahl
- Department of Physiology and Biophysics, University of Miami School of Medicine, Miami, Florida 33136, USA;
| |
Collapse
|
24
|
Güiza J, Barría I, Sáez JC, Vega JL. Innexins: Expression, Regulation, and Functions. Front Physiol 2018; 9:1414. [PMID: 30364195 PMCID: PMC6193117 DOI: 10.3389/fphys.2018.01414] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/18/2018] [Indexed: 01/02/2023] Open
Abstract
The innexin (Inx) proteins form gap junction channels and non-junctional channels (named hemichannels) in invertebrates. These channels participate in cellular communication playing a relevant role in several physiological processes. Pioneer studies conducted mainly in worms and flies have shown that innexins participate in embryo development and behavior. However, recent studies have elucidated new functions of innexins in Arthropoda, Nematoda, Annelida, and Cnidaria, such as immune response, and apoptosis. This review describes emerging data of possible new roles of innexins and summarizes the data available to date.
Collapse
Affiliation(s)
- Juan Güiza
- Laboratorio de Fisiología Experimental, Instituto Antofagasta, Universidad de Antofagasta, Antofagasta, Chile
| | - Iván Barría
- Laboratorio de Fisiología Experimental, Instituto Antofagasta, Universidad de Antofagasta, Antofagasta, Chile
| | - Juan C Sáez
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile.,Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - José L Vega
- Laboratorio de Fisiología Experimental, Instituto Antofagasta, Universidad de Antofagasta, Antofagasta, Chile
| |
Collapse
|
25
|
Molica F, Figueroa XF, Kwak BR, Isakson BE, Gibbins JM. Connexins and Pannexins in Vascular Function and Disease. Int J Mol Sci 2018; 19:ijms19061663. [PMID: 29874791 PMCID: PMC6032213 DOI: 10.3390/ijms19061663] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/28/2018] [Accepted: 05/31/2018] [Indexed: 12/24/2022] Open
Abstract
Connexins (Cxs) and pannexins (Panxs) are ubiquitous membrane channel forming proteins that are critically involved in many aspects of vascular physiology and pathology. The permeation of ions and small metabolites through Panx channels, Cx hemichannels and gap junction channels confers a crucial role to these proteins in intercellular communication and in maintaining tissue homeostasis. This review provides an overview of current knowledge with respect to the pathophysiological role of these channels in large arteries, the microcirculation, veins, the lymphatic system and platelet function. The essential nature of these membrane proteins in vascular homeostasis is further emphasized by the pathologies that are linked to mutations and polymorphisms in Cx and Panx genes.
Collapse
Affiliation(s)
- Filippo Molica
- Department of Pathology and Immunology, University of Geneva, CH-1211 Geneva, Switzerland.
| | - Xavier F Figueroa
- Departamento de Fisiología, Faculdad de Ciencias Biológicas, Pontifica Universidad Católica de Chile, Santiago 8330025, Chile.
| | - Brenda R Kwak
- Department of Pathology and Immunology, University of Geneva, CH-1211 Geneva, Switzerland.
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| | - Jonathan M Gibbins
- Institute for Cardiovascular & Metabolic Research, School of Biological Sciences, Harborne Building, University of Reading, Reading RG6 6AS, UK.
| |
Collapse
|
26
|
Romanov RA, Lasher RS, High B, Savidge LE, Lawson A, Rogachevskaja OA, Zhao H, Rogachevsky VV, Bystrova MF, Churbanov GD, Adameyko I, Harkany T, Yang R, Kidd GJ, Marambaud P, Kinnamon JC, Kolesnikov SS, Finger TE. Chemical synapses without synaptic vesicles: Purinergic neurotransmission through a CALHM1 channel-mitochondrial signaling complex. Sci Signal 2018; 11:11/529/eaao1815. [PMID: 29739879 DOI: 10.1126/scisignal.aao1815] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Conventional chemical synapses in the nervous system involve a presynaptic accumulation of neurotransmitter-containing vesicles, which fuse with the plasma membrane to release neurotransmitters that activate postsynaptic receptors. In taste buds, type II receptor cells do not have conventional synaptic features but nonetheless show regulated release of their afferent neurotransmitter, ATP, through a large-pore, voltage-gated channel, CALHM1. Immunohistochemistry revealed that CALHM1 was localized to points of contact between the receptor cells and sensory nerve fibers. Ultrastructural and super-resolution light microscopy showed that the CALHM1 channels were consistently associated with distinctive, large (1- to 2-μm) mitochondria spaced 20 to 40 nm from the presynaptic membrane. Pharmacological disruption of the mitochondrial respiratory chain limited the ability of taste cells to release ATP, suggesting that the immediate source of released ATP was the mitochondrion rather than a cytoplasmic pool of ATP. These large mitochondria may serve as both a reservoir of releasable ATP and the site of synthesis. The juxtaposition of the large mitochondria to areas of membrane displaying CALHM1 also defines a restricted compartment that limits the influx of Ca2+ upon opening of the nonselective CALHM1 channels. These findings reveal a distinctive organelle signature and functional organization for regulated, focal release of purinergic signals in the absence of synaptic vesicles.
Collapse
Affiliation(s)
- Roman A Romanov
- Institute of Cell Biophysics, Russian Academy of Science, Pushchino, Moscow Region 142290, Russia.,Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, A-1090 Vienna, Austria.,Immanuel Kant Baltic Federal University, Kaliningrad 236041, Russia
| | - Robert S Lasher
- Rocky Mountain Taste and Smell Center, Department of Cell and Developmental Biology, University Colorado School of Medicine, Aurora, CO 80045, USA
| | - Brigit High
- Rocky Mountain Taste and Smell Center, Department of Cell and Developmental Biology, University Colorado School of Medicine, Aurora, CO 80045, USA
| | - Logan E Savidge
- Rocky Mountain Taste and Smell Center, Department of Cell and Developmental Biology, University Colorado School of Medicine, Aurora, CO 80045, USA
| | - Adam Lawson
- Rocky Mountain Taste and Smell Center, Department of Cell and Developmental Biology, University Colorado School of Medicine, Aurora, CO 80045, USA
| | - Olga A Rogachevskaja
- Institute of Cell Biophysics, Russian Academy of Science, Pushchino, Moscow Region 142290, Russia
| | - Haitian Zhao
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | - Vadim V Rogachevsky
- Institute of Cell Biophysics, Russian Academy of Science, Pushchino, Moscow Region 142290, Russia.,United Pushchino Center for Electron Microscopy, Pushchino, Moscow Region 142290, Russia
| | - Marina F Bystrova
- Institute of Cell Biophysics, Russian Academy of Science, Pushchino, Moscow Region 142290, Russia
| | - Gleb D Churbanov
- Institute of Cell Biophysics, Russian Academy of Science, Pushchino, Moscow Region 142290, Russia
| | - Igor Adameyko
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, A-1090 Vienna, Austria.,Department of Physiology and Pharmacology, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, A-1090 Vienna, Austria.,Department of Neuroscience, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Ruibiao Yang
- Rocky Mountain Taste and Smell Center, Department of Cell and Developmental Biology, University Colorado School of Medicine, Aurora, CO 80045, USA
| | - Grahame J Kidd
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, and 3D-Electron Microscopy, Renovo Neural Inc., Cleveland, OH 44195, USA
| | - Philippe Marambaud
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | - John C Kinnamon
- Rocky Mountain Taste and Smell Center, Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| | - Stanislav S Kolesnikov
- Institute of Cell Biophysics, Russian Academy of Science, Pushchino, Moscow Region 142290, Russia.
| | - Thomas E Finger
- Rocky Mountain Taste and Smell Center, Department of Cell and Developmental Biology, University Colorado School of Medicine, Aurora, CO 80045, USA.
| |
Collapse
|
27
|
Volkenhoff A, Hirrlinger J, Kappel JM, Klämbt C, Schirmeier S. Live imaging using a FRET glucose sensor reveals glucose delivery to all cell types in the Drosophila brain. JOURNAL OF INSECT PHYSIOLOGY 2018; 106:55-64. [PMID: 28733241 DOI: 10.1016/j.jinsphys.2017.07.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 06/13/2017] [Accepted: 07/17/2017] [Indexed: 05/26/2023]
Abstract
All complex nervous systems are metabolically separated from circulation by a blood-brain barrier (BBB) that prevents uncontrolled leakage of solutes into the brain. Thus, all metabolites needed to sustain energy homeostasis must be transported across this BBB. In invertebrates, such as Drosophila, the major carbohydrate in circulation is the disaccharide trehalose and specific trehalose transporters are expressed by the glial BBB. Here we analyzed whether glucose is able to contribute to energy homeostasis in Drosophila. To study glucose influx into the brain we utilized a genetically encoded, FRET-based glucose sensor expressed in a cell type specific manner. When confronted with glucose all brain cells take up glucose within two minutes. In order to characterize the glucose transporter involved, we studied Drosophila Glut1, the homologue of which is primarily expressed by the BBB-forming endothelial cells and astrocytes in the mammalian nervous system. In Drosophila, however, Glut1 is expressed in neurons and is not found at the BBB. Thus, Glut1 cannot contribute to initial glucose uptake from the hemolymph. To test whether gap junctional coupling between the BBB forming cells and other neural cells contributes to glucose distribution we assayed these junctions using RNAi experiments and only found a minor contribution of gap junctions to glucose metabolism. Our results provide the entry point to further dissect the mechanisms underlying glucose distribution and offer new opportunities to understand brain metabolism.
Collapse
Affiliation(s)
- Anne Volkenhoff
- Institut für Neuro- und Verhaltensbiologie, Badestr. 9, 48149 Münster, Germany
| | - Johannes Hirrlinger
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, University of Leipzig, Liebigstr. 27, D-04103 Leipzig, Germany; Department of Neurogenetics, Max-Planck-Institute for Experimental Medicine, Hermann-Rein-Str. 3, D-37075 Göttingen, Germany
| | - Johannes M Kappel
- Institut für Neuro- und Verhaltensbiologie, Badestr. 9, 48149 Münster, Germany
| | - Christian Klämbt
- Institut für Neuro- und Verhaltensbiologie, Badestr. 9, 48149 Münster, Germany
| | - Stefanie Schirmeier
- Institut für Neuro- und Verhaltensbiologie, Badestr. 9, 48149 Münster, Germany.
| |
Collapse
|
28
|
Khaitin A, Rudkovskii M, Uzdensky A. Ca 2+ mediates axotomy-induced necrosis and apoptosis of satellite glial cells remote from the transection site in the isolated crayfish mechanoreceptor. Mol Cell Neurosci 2017; 88:7-15. [PMID: 29225183 DOI: 10.1016/j.mcn.2017.12.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 11/05/2017] [Accepted: 12/05/2017] [Indexed: 01/11/2023] Open
Abstract
Severe nerve injury such as axotomy induces neuron degeneration and death of surrounding glial cells. Using a crayfish stretch receptor that consists of a single mechanoreceptor neuron enveloped by satellite glia, we showed that axotomy not only mechanically injures glial cells at the transection location, but also induces necrosis or apoptosis of satellite glial cells remote from the transection site. We studied Ca2+role in spontaneous or axotomy-induced death of remote glial cells. Stretch receptors were isolated using the original technique that kept the neuron connected to the ventral cord ganglion (control preparations). Using Ca2+-sensitive fluorescence probe fluo-4, we showed Ca2+ accumulation in neuronal perikarion and glial envelope. Ca2+ gradually accumulated in glial cells after axotomy. In saline with triple Ca2+ concentration the axotomy-induced apoptosis of glial cells increased, but spontaneous or axotomy-induced necrosis was unexpectedly reduced. Saline with 1/3[Ca2+], oppositely, enhanced glial necrosis. Application of ionomycin, CdCl2, thapsigargin, and ryanodine showed the involvement of Ca2+ influx through ionic channels in the plasma membrane, inhibition of endoplasmic reticulum Ca2+-ATPase, and Ca2+ release from endoplasmic reticulum through ryanodine receptors in axotomy-induced glial necrosis. Apoptosis of glial cells surrounding axotomized neurons was promoted by ionomycin and thapsigargin. Possibly, other Ca2+ sources such as penetration through the plasma membrane contributed to axotomy-induced apoptosis and necrosis of remote glial cells. Thus, modulating different pathways that maintain calcium homeostasis, one can modulate axotomy-induced death of glial cells remote from the transection site.
Collapse
Affiliation(s)
- Andrey Khaitin
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don 344090, Russia
| | - Mikhail Rudkovskii
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don 344090, Russia
| | - Anatoly Uzdensky
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don 344090, Russia.
| |
Collapse
|
29
|
Zorzi V, Paciello F, Ziraldo G, Peres C, Mazzarda F, Nardin C, Pasquini M, Chiani F, Raspa M, Scavizzi F, Carrer A, Crispino G, Ciubotaru CD, Monyer H, Fetoni AR, M Salvatore A, Mammano F. Mouse Panx1 Is Dispensable for Hearing Acquisition and Auditory Function. Front Mol Neurosci 2017; 10:379. [PMID: 29234270 PMCID: PMC5712377 DOI: 10.3389/fnmol.2017.00379] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 10/30/2017] [Indexed: 11/13/2022] Open
Abstract
Panx1 forms plasma membrane channels in brain and several other organs, including the inner ear. Biophysical properties, activation mechanisms and modulators of Panx1 channels have been characterized in detail, however the impact of Panx1 on auditory function is unclear due to conflicts in published results. To address this issue, hearing performance and cochlear function of the Panx1−/− mouse strain, the first with a reported global ablation of Panx1, were scrutinized. Male and female homozygous (Panx1−/−), hemizygous (Panx1+/−) and their wild type (WT) siblings (Panx1+/+) were used for this study. Successful ablation of Panx1 was confirmed by RT-PCR and Western immunoblotting in the cochlea and brain of Panx1−/− mice. Furthermore, a previously validated Panx1-selective antibody revealed strong immunoreactivity in WT but not in Panx1−/− cochleae. Hearing sensitivity, outer hair cell-based “cochlear amplifier” and cochlear nerve function, analyzed by auditory brainstem response (ABR) and distortion product otoacoustic emission (DPOAE) recordings, were normal in Panx1+/− and Panx1−/− mice. In addition, we determined that global deletion of Panx1 impacts neither on connexin expression, nor on gap-junction coupling in the developing organ of Corti. Finally, spontaneous intercellular Ca2+ signal (ICS) activity in organotypic cochlear cultures, which is key to postnatal development of the organ of Corti and essential for hearing acquisition, was not affected by Panx1 ablation. Therefore, our results provide strong evidence that, in mice, Panx1 is dispensable for hearing acquisition and auditory function.
Collapse
Affiliation(s)
- Veronica Zorzi
- CNR Institute of Cell Biology and Neurobiology, Monterotondo, Italy.,School of Medicine, Institute of Otolaryngology, Catholic University, Rome, Italy
| | - Fabiola Paciello
- CNR Institute of Cell Biology and Neurobiology, Monterotondo, Italy
| | - Gaia Ziraldo
- CNR Institute of Cell Biology and Neurobiology, Monterotondo, Italy.,School of Medicine, Institute of Otolaryngology, Catholic University, Rome, Italy
| | - Chiara Peres
- CNR Institute of Cell Biology and Neurobiology, Monterotondo, Italy
| | - Flavia Mazzarda
- CNR Institute of Cell Biology and Neurobiology, Monterotondo, Italy.,Department of Science, Roma Tre University, Rome, Italy
| | - Chiara Nardin
- CNR Institute of Cell Biology and Neurobiology, Monterotondo, Italy.,Department of Science, Roma Tre University, Rome, Italy
| | - Miriam Pasquini
- CNR Institute of Cell Biology and Neurobiology, Monterotondo, Italy.,Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
| | - Francesco Chiani
- CNR Institute of Cell Biology and Neurobiology, Monterotondo, Italy
| | - Marcello Raspa
- CNR Institute of Cell Biology and Neurobiology, Monterotondo, Italy
| | | | - Andrea Carrer
- Department of Physics and Astronomy G. Galilei, University of Padua, Padua, Italy
| | - Giulia Crispino
- Department of Physics and Astronomy G. Galilei, University of Padua, Padua, Italy
| | | | - Hannah Monyer
- Department of Clinical Neurobiology, Deutches Krebforschungzentrum, University of Heidelberg, Heidelberg, Germany
| | - Anna R Fetoni
- School of Medicine, Institute of Otolaryngology, Catholic University, Rome, Italy
| | - Anna M Salvatore
- CNR Institute of Cell Biology and Neurobiology, Monterotondo, Italy
| | - Fabio Mammano
- CNR Institute of Cell Biology and Neurobiology, Monterotondo, Italy.,Department of Physics and Astronomy G. Galilei, University of Padua, Padua, Italy.,Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| |
Collapse
|
30
|
Ahn CS, Kim JG, Bae YA, Kim SH, Shin JH, Yang Y, Kang I, Kong Y. Fasciclin-calcareous corpuscle binary complex mediated protein-protein interactions in Taenia solium metacestode. Parasit Vectors 2017; 10:438. [PMID: 28931431 PMCID: PMC5606126 DOI: 10.1186/s13071-017-2359-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 08/31/2017] [Indexed: 01/30/2023] Open
Abstract
Background Neurocysticercosis (NC) caused by Taenia solium metacestode (TsM) is a serious neurological disease of global concern. Diverse bioactive molecules involved in the long-term survival of TsM might contribute to disease progression. Fasciclin (Fas) is an extracellular protein that mediates adhesion, migration and differentiation of cells by interacting with other molecules. We hypothesized that TsMFas might bind to calcareous corpuscle (CC) through its adhesive property and participate in crucial protein-protein interactions, thus contributing to the creation of a symbiotic interactome network. Methods Two paralogous TsMFas (TsMFas1 and TsMFas2) were isolated, and their molecular properties were characterized. The co-localization pattern of TsMFas1 and TsMFas2 with CC was determined. CC-TsMFas binary complex was generated by incubating CC with recombinant proteins (rTsMFas1 and 2). In vitro binding assay of CC-rTsMFas1 or CC-rTsMFas2 binary complex with TsM cellular proteins extracted from scolex and neck was conducted. Their binding partners were identified through proteomic analysis. Integrated protein-protein interaction networks were established. Results TsMFas1 (6072 bp long) was composed of 15 exons (841 amino acid polypeptide) interrupted by 14 introns. TsMFas2 (5201 bp long) comprised of 11 exons (597 amino acids) and 10 intervening introns. These proteins displayed 22% amino acid sequence identity to each other, but tightly conserved Fas-related domains. Several isoforms of Fas1 and Fas2 proteins might have been expressed through post-translational modifications. They showed adhesion activity with other cells. TsMFas proteins were largely distributed in parenchymal regions of the scolex and bladder wall. These molecules were co-localized with CC, a unique organelle found in platyhelminths. Subsequent proteome analysis of CC-Fas binary complex mediated protein-protein interactions revealed seven protein ligands in the TsM cellular proteins. Their functions were mainly segregated into carbohydrate metabolism (enolase, phosphoenolpyruvate carboxykinase, phosphoglycerate kinase and glyceraldehyde 3-phosphate dehydrogenase) and cytoskeleton/cellular motility (actin, paramyosin and innexin nuc-9). Those proteins had direct (physical) and/or indirect (functional) relationships along with their biochemical properties and biological roles. Conclusion Protein repertoires strongly suggest that TsMFas and CC may symbiotically mediate protein-protein interactions during biological processes to maintain efficacious homeostatic functions and ensure the prolonged survival of TsM in the host. Electronic supplementary material The online version of this article (10.1186/s13071-017-2359-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chun-Seob Ahn
- Department of Molecular Parasitology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea
| | - Jeong-Geun Kim
- Department of Molecular Parasitology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea
| | - Young-An Bae
- Department of Microbiology, Gachon University College of Medicine, Incheon, 21936, South Korea
| | - Seon-Hee Kim
- Department of Microbiology, Gachon University College of Medicine, Incheon, 21936, South Korea
| | - Joo-Ho Shin
- Division of Pharmacology, Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea
| | - Yichao Yang
- Guangxi Centers for Disease Prevention and Control, Nanning, Guangxi, 53002, China
| | - Insug Kang
- Department of Molecular Biology and Biochemistry, School of Medicine, Kyung Hee University, Seoul, 02447, South Korea
| | - Yoon Kong
- Department of Molecular Parasitology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea.
| |
Collapse
|
31
|
Whyte-Fagundes P, Zoidl G. Mechanisms of pannexin1 channel gating and regulation. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:65-71. [PMID: 28735901 DOI: 10.1016/j.bbamem.2017.07.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 07/17/2017] [Accepted: 07/18/2017] [Indexed: 01/07/2023]
Abstract
Pannexins are a family of integral membrane proteins with distinct post-translational modifications, sub-cellular localization and tissue distribution. Panx1 is the most studied and best-characterized isoform of this gene family. The ubiquitous expression, as well as its function as a major ATP release and nucleotide permeation channel, makes Panx1 a primary candidate for participating in the pathophysiology of CNS disorders. While many investigations revolve around Panx1 functions in health and disease, more recently, details started emerging about mechanisms that control Panx1 channel activity. These advancements in Panx1 biology have revealed that beyond its classical role as an unopposed plasma membrane channel, it participates in alternative pathways involving multiple intracellular compartments, protein complexes and a myriad of extracellular participants. Here, we review recent progress in our understanding of Panx1 at the center of these pathways, highlighting its modulation in a context specific manner. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
| | - Georg Zoidl
- Biology, York University, Toronto, Canada; Psychology, York University, Toronto, Canada.
| |
Collapse
|
32
|
Otopalik AG, Lane B, Schulz DJ, Marder E. Innexin expression in electrically coupled motor circuits. Neurosci Lett 2017; 695:19-24. [PMID: 28711343 PMCID: PMC5767152 DOI: 10.1016/j.neulet.2017.07.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 06/22/2017] [Accepted: 07/11/2017] [Indexed: 12/21/2022]
Abstract
The many roles of innexins, the molecules that form gap junctions in invertebrates, have been explored in numerous species. Here, we present a summary of innexin expression and function in two small, central pattern generating circuits found in crustaceans: the stomatogastric ganglion and the cardiac ganglion. The two ganglia express multiple innexin genes, exhibit varying combinations of symmetrical and rectifying gap junctions, as well as gap junctions within and across different cell types. Past studies have revealed correlations in ion channel and innexin expression in coupled neurons, as well as intriguing functional relationships between ion channel conductances and electrical coupling. Together, these studies suggest a putative role for innexins in correlating activity between coupled neurons at the levels of gene expression and physiological activity during development and in the adult animal.
Collapse
Affiliation(s)
- Adriane G Otopalik
- Volen Center and Biology Department, Brandeis University, Waltham, MA 02454, USA.
| | - Brian Lane
- Division of Biological Sciences, University of Missouri-Columbia, Columbia, MO, 65211, USA
| | - David J Schulz
- Division of Biological Sciences, University of Missouri-Columbia, Columbia, MO, 65211, USA
| | - Eve Marder
- Volen Center and Biology Department, Brandeis University, Waltham, MA 02454, USA
| |
Collapse
|
33
|
Skerrett IM, Williams JB. A structural and functional comparison of gap junction channels composed of connexins and innexins. Dev Neurobiol 2017; 77:522-547. [PMID: 27582044 PMCID: PMC5412853 DOI: 10.1002/dneu.22447] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 08/12/2016] [Accepted: 08/30/2016] [Indexed: 02/03/2023]
Abstract
Methods such as electron microscopy and electrophysiology led to the understanding that gap junctions were dense arrays of channels connecting the intracellular environments within almost all animal tissues. The characteristics of gap junctions were remarkably similar in preparations from phylogenetically diverse animals such as cnidarians and chordates. Although few studies directly compared them, minor differences were noted between gap junctions of vertebrates and invertebrates. For instance, a slightly wider gap was noted between cells of invertebrates and the spacing between invertebrate channels was generally greater. Connexins were identified as the structural component of vertebrate junctions in the 1980s and innexins as the structural component of pre-chordate junctions in the 1990s. Despite a lack of similarity in gene sequence, connexins and innexins are remarkably similar. Innexins and connexins have the same membrane topology and form intercellular channels that play a variety of tissue- and temporally specific roles. Both protein types oligomerize to form large aqueous channels that allow the passage of ions and small metabolites and are regulated by factors such as pH, calcium, and voltage. Much more is currently known about the structure, function, and structure-function relationships of connexins. However, the innexin field is expanding. Greater knowledge of innexin channels will permit more detailed comparisons with their connexin-based counterparts, and provide insight into the ubiquitous yet specific roles of gap junctions. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 522-547, 2017.
Collapse
Affiliation(s)
- I Martha Skerrett
- Biology Department, SUNY Buffalo State, 1300 Elmwood Ave, Buffalo, New York, 14222
| | - Jamal B Williams
- Biology Department, SUNY Buffalo State, 1300 Elmwood Ave, Buffalo, New York, 14222
| |
Collapse
|
34
|
Molica F, Stierlin FB, Fontana P, Kwak BR. Pannexin- and Connexin-Mediated Intercellular Communication in Platelet Function. Int J Mol Sci 2017; 18:E850. [PMID: 28420171 PMCID: PMC5412434 DOI: 10.3390/ijms18040850] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/06/2017] [Accepted: 04/12/2017] [Indexed: 12/11/2022] Open
Abstract
The three major blood cell types, i.e., platelets, erythrocytes and leukocytes, are all produced in the bone marrow. While red blood cells are the most numerous and white cells are the largest, platelets are small fragments and account for a minor part of blood volume. However, platelets display a crucial function by preventing bleeding. Upon vessel wall injury, platelets adhere to exposed extracellular matrix, become activated, and form a platelet plug preventing hemorrhagic events. However, when platelet activation is exacerbated, as in rupture of an atherosclerotic plaque, the same mechanism may lead to acute thrombosis causing major ischemic events such as myocardial infarction or stroke. In the past few years, major progress has been made in understanding of platelet function modulation. In this respect, membrane channels formed by connexins and/or pannexins are of particular interest. While it is still not completely understood whether connexins function as hemichannels or gap junction channels to inhibit platelet aggregation, there is clear-cut evidence for a specific implication of pannexin1 channels in collagen-induced aggregation. The focus of this review is to summarize current knowledge of the role of connexins and pannexins in platelet aggregation and to discuss possible pharmacological approaches along with their limitations and future perspectives for new potential therapies.
Collapse
Affiliation(s)
- Filippo Molica
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland.
- Department of Medical Specializations, Cardiology, University of Geneva, 1211 Geneva, Switzerland.
| | - Florian B Stierlin
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland.
- Department of Medical Specializations, Cardiology, University of Geneva, 1211 Geneva, Switzerland.
- Geneva Platelet Group, University of Geneva, 1211 Geneva, Switzerland.
| | - Pierre Fontana
- Geneva Platelet Group, University of Geneva, 1211 Geneva, Switzerland.
- Division of Angiology and Haemostasis, Geneva University Hospitals, 1211 Geneva, Switzerland.
| | - Brenda R Kwak
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland.
- Department of Medical Specializations, Cardiology, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
35
|
Hasegawa DK, Erickson SL, Hersh BM, Turnbull MW. Virus Innexins induce alterations in insect cell and tissue function. JOURNAL OF INSECT PHYSIOLOGY 2017; 98:173-181. [PMID: 28077262 DOI: 10.1016/j.jinsphys.2017.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 10/10/2016] [Accepted: 01/06/2017] [Indexed: 06/06/2023]
Abstract
Polydnaviruses are dsDNA viruses that induce immune and developmental alterations in their caterpillar hosts. Characterization of polydnavirus gene families and family members is necessary to understand mechanisms of pathology and evolution of these viruses, and may aid to elucidate the role of host homologues if present. For example, the polydnavirus vinnexin gene family encodes homologues of insect gap junction genes (innexins) that are expressed in host immune cells (hemocytes). While the roles of Innexin proteins and gap junctions in insect immunity are largely unclear, we previously demonstrated that Vinnexins form functional gap junctions and alter the junctional characteristics of a host Innexin when co-expressed in paired Xenopus oocytes. Here, we test the effect of ectopic vinnexin expression on host cell physiology using both a lepidopteran cell culture model and a dipteran whole organism model. Vinnexin expression in the cell culture system resulted in gene-specific alterations in cell morphology and a slight, but non-statistically significant, reduction in gap junction activity as measured by dye transfer, while ectopic expression of a lepidopteran innexin2 gene led to morphological alterations and increase in gap junction activity. Global ectopic expression in the model dipteran, Drosophila melanogaster, of one vinnexin (vinnexinG) or D. melanogaster innexin2 (Dm-inx2) resulted in embryonic lethality, while expression of the other vinnexin genes had no effect. Furthermore, ectopic expression of vinnexinG, but not other vinnexin genes or Dm-inx2, in D. melanogaster larval gut resulted in developmental arrest in the pupal stage. These data indicate the vinnexins likely have gene-specific roles in host manipulation. They also support the use of Drosophila in further analysis of the role of Vinnexins and other polydnavirus genes in modifying host physiological processes. Finally, our findings suggest the vinnexin genes may be useful to perturb and characterize the physiological functions of insect Innexins.
Collapse
Affiliation(s)
- Daniel K Hasegawa
- Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA.
| | | | - Bradley M Hersh
- Department of Biology, Allegheny College, Meadville, PA 16335, USA.
| | - Matthew W Turnbull
- Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA; Department of Plant and Environmental Sciences, Clemson University, Clemson, SC 29634, USA.
| |
Collapse
|
36
|
Burrell BD. Comparative biology of pain: What invertebrates can tell us about how nociception works. J Neurophysiol 2017; 117:1461-1473. [PMID: 28053241 DOI: 10.1152/jn.00600.2016] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 01/04/2017] [Accepted: 01/04/2017] [Indexed: 12/30/2022] Open
Abstract
The inability to adequately treat chronic pain is a worldwide health care crisis. Pain has both an emotional and a sensory component, and this latter component, nociception, refers specifically to the detection of damaging or potentially damaging stimuli. Nociception represents a critical interaction between an animal and its environment and exhibits considerable evolutionary conservation across species. Using comparative approaches to understand the basic biology of nociception could promote the development of novel therapeutic strategies to treat pain, and studies of nociception in invertebrates can provide especially useful insights toward this goal. Both vertebrates and invertebrates exhibit segregated sensory pathways for nociceptive and nonnociceptive information, injury-induced sensitization to nociceptive and nonnociceptive stimuli, and even similar antinociceptive modulatory processes. In a number of invertebrate species, the central nervous system is understood in considerable detail, and it is often possible to record from and/or manipulate single identifiable neurons through either molecular genetic or physiological approaches. Invertebrates also provide an opportunity to study nociception in an ethologically relevant context that can provide novel insights into the nature of how injury-inducing stimuli produce persistent changes in behavior. Despite these advantages, invertebrates have been underutilized in nociception research. In this review, findings from invertebrate nociception studies are summarized, and proposals for how research using invertebrates can address questions about the fundamental mechanisms of nociception are presented.
Collapse
Affiliation(s)
- Brian D Burrell
- Division of Basic Biomedical Sciences, Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| |
Collapse
|
37
|
Characterization of the Tetraspan Junctional Complex (4JC) superfamily. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1859:402-414. [PMID: 27916633 DOI: 10.1016/j.bbamem.2016.11.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 11/22/2016] [Accepted: 11/29/2016] [Indexed: 12/31/2022]
Abstract
Connexins or innexins form gap junctions, while claudins and occludins form tight junctions. In this study, statistical data, derived using novel software, indicate that these four junctional protein families and eleven other families of channel and channel auxiliary proteins are related by common descent and comprise the Tetraspan (4 TMS) Junctional Complex (4JC) Superfamily. These proteins all share similar 4 transmembrane α-helical (TMS) topologies. Evidence is presented that they arose via an intragenic duplication event, whereby a 2 TMS-encoding genetic element duplicated tandemly to give 4 TMS proteins. In cases where high resolution structural data were available, the conclusion of homology was supported by conducting structural comparisons. Phylogenetic trees reveal the probable relationships of these 15 families to each other. Long homologues containing fusions to other recognizable domains as well as internally duplicated or fused domains are reported. Large "fusion" proteins containing 4JC domains proved to fall predominantly into family-specific patterns as follows: (1) the 4JC domain was N-terminal; (2) the 4JC domain was C-terminal; (3) the 4JC domain was duplicated or occasionally triplicated and (4) mixed fusion types were present. Our observations provide insight into the evolutionary origins and subfunctions of these proteins as well as guides concerning their structural and functional relationships.
Collapse
|
38
|
Cronstein BN, Sitkovsky M. Adenosine and adenosine receptors in the pathogenesis and treatment of rheumatic diseases. Nat Rev Rheumatol 2016; 13:41-51. [PMID: 27829671 DOI: 10.1038/nrrheum.2016.178] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Adenosine, a nucleoside derived primarily from the extracellular hydrolysis of adenine nucleotides, is a potent regulator of inflammation. Adenosine mediates its effects on inflammatory cells by engaging one or more cell-surface receptors. The expression and function of adenosine receptors on different cell types change during the course of rheumatic diseases, such as rheumatoid arthritis (RA). Targeting adenosine receptors directly for the treatment of rheumatic diseases is currently under study; however, indirect targeting of adenosine receptors by enhancing adenosine levels at inflamed sites accounts for most of the anti-inflammatory effects of methotrexate, the anchor drug for the treatment of RA. In this Review, we discuss the regulation of extracellular adenosine levels and the role of adenosine in regulating the inflammatory and immune responses in rheumatic diseases such as RA, psoriasis and other types of inflammatory arthritis. In addition, adenosine and its receptors are involved in promoting fibrous matrix production in the skin and other organs, and the role of adenosine in fibrosis and fibrosing diseases is also discussed.
Collapse
Affiliation(s)
- Bruce N Cronstein
- NYU-HHC Clinical and Translational Science Institute, NYU School of Medicine, 550 First Avenue, New York, New York 10016, USA
| | - Michail Sitkovsky
- New England Inflammation and Tissue Protection Institute, Northeastern University, 360 Huntington Avenue, 312 MU, Boston, Massachusetts 02115, USA
| |
Collapse
|
39
|
Lautemann J, Bohrmann J. Relating proton pumps with gap junctions: colocalization of ductin, the channel-forming subunit c of V-ATPase, with subunit a and with innexins 2 and 3 during Drosophila oogenesis. BMC DEVELOPMENTAL BIOLOGY 2016; 16:24. [PMID: 27412523 PMCID: PMC4944501 DOI: 10.1186/s12861-016-0124-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 06/29/2016] [Indexed: 01/21/2023]
Abstract
BACKGROUND Ion-transport mechanisms and gap junctions are known to cooperate in creating bioelectric phenomena, like pH gradients, voltage gradients and ion fluxes within single cells, tissues, organs, and whole organisms. Such phenomena have been shown to play regulatory roles in a variety of developmental and regenerative processes. Using Drosophila oogenesis as a model system, we aim at characterizing in detail the mechanisms underlying bioelectric phenomena in order to reveal their regulatory functions. We, therefore, investigated the stage-specific distribution patterns of V-ATPase components in relation to gap-junction proteins. RESULTS We analysed the localization of the V-ATPase components ductin (subunit c) and subunit a, and the gap-junction components innexins 2 and 3, especially in polar cells, border cells, stalk cells and centripetally migrating cells. These types of follicle cells had previously been shown to exhibit characteristic patterns of membrane channels as well as membrane potential and intracellular pH. Stage-specifically, ductin and subunit a were found either colocalized or separately enriched in different regions of soma and germ-line cells. While ductin was often more prominent in plasma membranes, subunit a was more prominent in cytoplasmic and nuclear vesicles. Particularly, ductin was enriched in polar cells, stalk cells, and nurse-cell membranes, whereas subunit a was enriched in the cytoplasm of border cells, columnar follicle cells and germ-line cells. Comparably, ductin and both innexins 2 and 3 were either colocalized or separately enriched in different cellular regions. While ductin often showed a continuous membrane distribution, the distribution of both innexins was mostly punctate. Particularly, ductin was enriched in polar cells and stalk cells, whereas innexin 2 was enriched in the oolemma, and innexin 3 in centripetally migrating follicle cells. In lateral follicle-cell membranes, the three proteins were found colocalized as well as separately concentrated in presumed gap-junction plaques. CONCLUSIONS Our results support the notion of a large variety of gap junctions existing in the Drosophila ovary. Moreover, since ductin is the channel-forming part of a proton pump and, like the innexins, is able to form junctional as well as non-junctional membrane channels, a plethora of cellular functions could be realized by using these proteins. The distribution and activity patterns of such membrane channels are expected to contribute to developmentally important bioelectric signals.
Collapse
Affiliation(s)
- Julia Lautemann
- Institut für Biologie II, RWTH Aachen University, Abt. Zoologie und Humanbiologie, Worringerweg 3, 52056, Aachen, Germany
| | - Johannes Bohrmann
- Institut für Biologie II, RWTH Aachen University, Abt. Zoologie und Humanbiologie, Worringerweg 3, 52056, Aachen, Germany.
| |
Collapse
|
40
|
Zemkova H, Tomić M, Kucka M, Aguilera G, Stojilkovic SS. Spontaneous and CRH-Induced Excitability and Calcium Signaling in Mice Corticotrophs Involves Sodium, Calcium, and Cation-Conducting Channels. Endocrinology 2016; 157:1576-89. [PMID: 26901094 PMCID: PMC4816721 DOI: 10.1210/en.2015-1899] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Transgenic mice expressing the tdimer2(12) form of Discosoma red fluorescent protein under control of the proopiomelanocortin gene's regulatory elements are a useful model for studying corticotrophs. Using these mice, we studied the ion channels and mechanisms controlling corticotroph excitability. Corticotrophs were either quiescent or electrically active, with a 22-mV difference in the resting membrane potential (RMP) between the 2 groups. In quiescent cells, CRH depolarized the membrane, leading to initial single spiking and sustained bursting; in active cells, CRH further facilitated or inhibited electrical activity and calcium spiking, depending on the initial activity pattern and CRH concentration. The stimulatory but not inhibitory action of CRH on electrical activity was mimicked by cAMP independently of the presence or absence of arachidonic acid. Removal of bath sodium silenced spiking and hyperpolarized the majority of cells; in contrast, the removal of bath calcium did not affect RMP but reduced CRH-induced depolarization, which abolished bursting electrical activity and decreased the spiking frequency but not the amplitude of single spikes. Corticotrophs with inhibited voltage-gated sodium channels fired calcium-dependent action potentials, whereas cells with inhibited L-type calcium channels fired sodium-dependent spikes; blockade of both channels abolished spiking without affecting the RMP. These results indicate that the background voltage-insensitive sodium conductance influences RMP, the CRH-depolarization current is driven by a cationic conductance, and the interplay between voltage-gated sodium and calcium channels plays a critical role in determining the status and pattern of electrical activity and calcium signaling.
Collapse
Affiliation(s)
- Hana Zemkova
- Department of Cellular and Molecular Neuroendocrinology, Institute of Physiology Academy of Sciences of the Czech Republic, 142 20 Prague 4, Czech Republic (H.Z.); and Sections on Cellular Signaling (H.Z., M.T., M.K., S.S.S.) and Endocrine Physiology (G.A.), Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892-4510
| | - Melanija Tomić
- Department of Cellular and Molecular Neuroendocrinology, Institute of Physiology Academy of Sciences of the Czech Republic, 142 20 Prague 4, Czech Republic (H.Z.); and Sections on Cellular Signaling (H.Z., M.T., M.K., S.S.S.) and Endocrine Physiology (G.A.), Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892-4510
| | - Marek Kucka
- Department of Cellular and Molecular Neuroendocrinology, Institute of Physiology Academy of Sciences of the Czech Republic, 142 20 Prague 4, Czech Republic (H.Z.); and Sections on Cellular Signaling (H.Z., M.T., M.K., S.S.S.) and Endocrine Physiology (G.A.), Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892-4510
| | - Greti Aguilera
- Department of Cellular and Molecular Neuroendocrinology, Institute of Physiology Academy of Sciences of the Czech Republic, 142 20 Prague 4, Czech Republic (H.Z.); and Sections on Cellular Signaling (H.Z., M.T., M.K., S.S.S.) and Endocrine Physiology (G.A.), Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892-4510
| | - Stanko S Stojilkovic
- Department of Cellular and Molecular Neuroendocrinology, Institute of Physiology Academy of Sciences of the Czech Republic, 142 20 Prague 4, Czech Republic (H.Z.); and Sections on Cellular Signaling (H.Z., M.T., M.K., S.S.S.) and Endocrine Physiology (G.A.), Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892-4510
| |
Collapse
|
41
|
Oliveira FM, Coelho IEV, Lopes MD, Taranto AG, Junior MC, Santos LLD, Villar JAPF, Fonseca CT, Lopes DDO. The Use of Reverse Vaccinology and Molecular Modeling Associated with Cell Proliferation Stimulation Approach to Select Promiscuous Epitopes from Schistosoma mansoni. Appl Biochem Biotechnol 2016; 179:1023-40. [DOI: 10.1007/s12010-016-2048-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 03/07/2016] [Indexed: 12/11/2022]
|
42
|
Dahl G. ATP release through pannexon channels. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0191. [PMID: 26009770 DOI: 10.1098/rstb.2014.0191] [Citation(s) in RCA: 179] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Extracellular adenosine triphosphate (ATP) serves as a signal for diverse physiological functions, including spread of calcium waves between astrocytes, control of vascular oxygen supply and control of ciliary beat in the airways. ATP can be released from cells by various mechanisms. This review focuses on channel-mediated ATP release and its main enabler, Pannexin1 (Panx1). Six subunits of Panx1 form a plasma membrane channel termed 'pannexon'. Depending on the mode of stimulation, the pannexon has large conductance (500 pS) and unselective permeability to molecules less than 1.5 kD or is a small (50 pS), chloride-selective channel. Most physiological and pathological stimuli induce the large channel conformation, whereas the small conformation so far has only been observed with exclusive voltage activation of the channel. The interaction between pannexons and ATP is intimate. The pannexon is not only the conduit for ATP, permitting ATP efflux from cells down its concentration gradient, but the pannexon is also modulated by ATP. The channel can be activated by ATP through both ionotropic P2X as well as metabotropic P2Y purinergic receptors. In the absence of a control mechanism, this positive feedback loop would lead to cell death owing to the linkage of purinergic receptors with apoptotic processes. A control mechanism preventing excessive activation of the purinergic receptors is provided by ATP binding (with low affinity) to the Panx1 protein and gating the channel shut.
Collapse
Affiliation(s)
- Gerhard Dahl
- School of Medicine, University of Miami, 1600 NW 10th Avenue, Miami, FL 33136, USA
| |
Collapse
|
43
|
Marsh A, Casey-Green K, Probert F, Withall D, Mitchell DA, Dilly SJ, James S, Dimitri W, Ladwa SR, Taylor PC, Singer DRJ. Simvastatin Sodium Salt and Fluvastatin Interact with Human Gap Junction Gamma-3 Protein. PLoS One 2016; 11:e0148266. [PMID: 26863535 PMCID: PMC4749215 DOI: 10.1371/journal.pone.0148266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 01/15/2016] [Indexed: 11/26/2022] Open
Abstract
Finding pleiomorphic targets for drugs allows new indications or warnings for treatment to be identified. As test of concept, we applied a new chemical genomics approach to uncover additional targets for the widely prescribed lipid-lowering pro-drug simvastatin. We used mRNA extracted from internal mammary artery from patients undergoing coronary artery surgery to prepare a viral cardiovascular protein library, using T7 bacteriophage. We then studied interactions of clones of the bacteriophage, each expressing a different cardiovascular polypeptide, with surface-bound simvastatin in 96-well plates. To maximise likelihood of identifying meaningful interactions between simvastatin and vascular peptides, we used a validated photo-immobilisation method to apply a series of different chemical linkers to bind simvastatin so as to present multiple orientations of its constituent components to potential targets. Three rounds of biopanning identified consistent interaction with the clone expressing part of the gene GJC3, which maps to Homo sapiens chromosome 7, and codes for gap junction gamma-3 protein, also known as connexin 30.2/31.3 (mouse connexin Cx29). Further analysis indicated the binding site to be for the N-terminal domain putatively 'regulating' connexin hemichannel and gap junction pores. Using immunohistochemistry we found connexin 30.2/31.3 to be present in samples of artery similar to those used to prepare the bacteriophage library. Surface plasmon resonance revealed that a 25 amino acid synthetic peptide representing the discovered N-terminus did not interact with simvastatin lactone, but did bind to the hydrolysed HMG CoA inhibitor, simvastatin acid. This interaction was also seen for fluvastatin. The gap junction blockers carbenoxolone and flufenamic acid also interacted with the same peptide providing insight into potential site of binding. These findings raise key questions about the functional significance of GJC3 transcripts in the vasculature and other tissues, and this connexin's role in therapeutic and adverse effects of statins in a range of disease states.
Collapse
Affiliation(s)
- Andrew Marsh
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | | | - Fay Probert
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - David Withall
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - Daniel A. Mitchell
- Division of Metabolic and Vascular Health, Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, Coventry, CV2 2DX, United Kingdom
| | - Suzanne J. Dilly
- Tangent Reprofiling Ltd, c/o SEEK, Central Point, 45 Beech Street, London, EC2Y 8AD, United Kingdom
| | - Sean James
- University Hospital Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, United Kingdom
| | - Wade Dimitri
- University Hospital Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, United Kingdom
| | - Sweta R. Ladwa
- Tangent Reprofiling Ltd, c/o SEEK, Central Point, 45 Beech Street, London, EC2Y 8AD, United Kingdom
| | - Paul C. Taylor
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - Donald R. J. Singer
- University Hospital Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, United Kingdom
- Fellowship of Postgraduate Medicine, 11 Chandos St, London W1G 9EB, United Kingdom
- Division of Metabolic and Vascular Health, Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, Coventry, CV2 2DX, United Kingdom
| |
Collapse
|
44
|
Moroz LL, Kohn AB. Independent origins of neurons and synapses: insights from ctenophores. Philos Trans R Soc Lond B Biol Sci 2016; 371:20150041. [PMID: 26598724 PMCID: PMC4685580 DOI: 10.1098/rstb.2015.0041] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2015] [Indexed: 12/29/2022] Open
Abstract
There is more than one way to develop neuronal complexity, and animals frequently use different molecular toolkits to achieve similar functional outcomes. Genomics and metabolomics data from basal metazoans suggest that neural signalling evolved independently in ctenophores and cnidarians/bilaterians. This polygenesis hypothesis explains the lack of pan-neuronal and pan-synaptic genes across metazoans, including remarkable examples of lineage-specific evolution of neurogenic and signalling molecules as well as synaptic components. Sponges and placozoans are two lineages without neural and muscular systems. The possibility of secondary loss of neurons and synapses in the Porifera/Placozoa clades is a highly unlikely and less parsimonious scenario. We conclude that acetylcholine, serotonin, histamine, dopamine, octopamine and gamma-aminobutyric acid (GABA) were recruited as transmitters in the neural systems in cnidarian and bilaterian lineages. By contrast, ctenophores independently evolved numerous secretory peptides, indicating extensive adaptations within the clade and suggesting that early neural systems might be peptidergic. Comparative analysis of glutamate signalling also shows numerous lineage-specific innovations, implying the extensive use of this ubiquitous metabolite and intercellular messenger over the course of convergent and parallel evolution of mechanisms of intercellular communication. Therefore: (i) we view a neuron as a functional character but not a genetic character, and (ii) any given neural system cannot be considered as a single character because it is composed of different cell lineages with distinct genealogies, origins and evolutionary histories. Thus, when reconstructing the evolution of nervous systems, we ought to start with the identification of particular cell lineages by establishing distant neural homologies or examples of convergent evolution. In a corollary of the hypothesis of the independent origins of neurons, our analyses suggest that both electrical and chemical synapses evolved more than once.
Collapse
Affiliation(s)
- Leonid L Moroz
- The Whitney Laboratory for Marine Bioscience, 9505 Ocean Shore Boulevard, St Augustine, FL 32080, USA Department of Neuroscience and McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Andrea B Kohn
- The Whitney Laboratory for Marine Bioscience, 9505 Ocean Shore Boulevard, St Augustine, FL 32080, USA
| |
Collapse
|
45
|
von Bernhardi R, Eugenín-von Bernhardi J, Flores B, Eugenín León J. Glial Cells and Integrity of the Nervous System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 949:1-24. [PMID: 27714682 DOI: 10.1007/978-3-319-40764-7_1] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Today, there is enormous progress in understanding the function of glial cells, including astroglia, oligodendroglia, Schwann cells, and microglia. Around 150 years ago, glia were viewed as a glue among neurons. During the course of the twentieth century, microglia were discovered and neuroscientists' views evolved toward considering glia only as auxiliary cells of neurons. However, over the last two to three decades, glial cells' importance has been reconsidered because of the evidence on their involvement in defining central nervous system architecture, brain metabolism, the survival of neurons, development and modulation of synaptic transmission, propagation of nerve impulses, and many other physiological functions. Furthermore, increasing evidence shows that glia are involved in the mechanisms of a broad spectrum of pathologies of the nervous system, including some psychiatric diseases, epilepsy, and neurodegenerative diseases to mention a few. It appears safe to say that no neurological disease can be understood without considering neuron-glia crosstalk. Thus, this book aims to show different roles played by glia in the healthy and diseased nervous system, highlighting some of their properties while considering that the various glial cell types are essential components not only for cell function and integration among neurons, but also for the emergence of important brain homeostasis.
Collapse
Affiliation(s)
- Rommy von Bernhardi
- Department of Neurology, School of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile.
| | - Jaime Eugenín-von Bernhardi
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University, Pettenkoferstr.12, 80336, Munich, Germany.,Graduate School of Systemic Neuroscience, Ludwig-Maximilians-University, 82152, Planegg-Martinsried, Munich, Germany
| | - Betsi Flores
- Department of Neurology, School of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| | - Jaime Eugenín León
- Department of Biology, Faculty of Chemistry and Biology, USACH, Santiago, Chile
| |
Collapse
|
46
|
Abstract
The pancreas produces enzymes with a digestive function and hormones with a metabolic function, which are produced by distinct cell types of acini and islets, respectively. Within these units, secretory cells coordinate their functioning by exchanging information via signals that flow in the intercellular spaces and are generated either at distance (several neural and hormonal inputs) or nearby the pancreatic cells themselves (inputs mediated by membrane ionic-specific channels and by ionic- and metabolite-permeant pannexin channels and connexin "hemichannels"). Pancreatic secretory cells further interact via the extracellular matrix of the pancreas (inputs mediated by integrins) and directly with neighboring cells, by mechanisms that do not require extracellular mediators (inputs mediated by gap and tight junction channels). Here, we review the expression and function of the connexins and pannexins that are expressed by the main secretory cells of the exocrine and endocrine pancreatic cells. Available data show that the patterns of expression of these proteins differ in acini and islets, supporting distinct functions in the physiological secretion of pancreatic enzymes and hormones. Circumstantial evidence further suggests that alterations in the signaling provided by these proteins are involved in pancreatic diseases.
Collapse
|
47
|
Bowie D, Attwell D. Coupling cellular metabolism to neuronal signalling. J Physiol 2015; 593:3413-5. [PMID: 26272625 DOI: 10.1113/jp271075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 06/19/2015] [Indexed: 01/15/2023] Open
Affiliation(s)
- Derek Bowie
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada, H3G 1Y6
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK
| |
Collapse
|
48
|
Decrock E, De Bock M, Wang N, Bultynck G, Giaume C, Naus CC, Green CR, Leybaert L. Connexin and pannexin signaling pathways, an architectural blueprint for CNS physiology and pathology? Cell Mol Life Sci 2015; 72:2823-51. [PMID: 26118660 PMCID: PMC11113968 DOI: 10.1007/s00018-015-1962-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 06/11/2015] [Indexed: 02/06/2023]
Abstract
The central nervous system (CNS) is composed of a highly heterogeneous population of cells. Dynamic interactions between different compartments (neuronal, glial, and vascular systems) drive CNS function and allow to integrate and process information as well as to respond accordingly. Communication within this functional unit, coined the neuro-glio-vascular unit (NGVU), typically relies on two main mechanisms: direct cell-cell coupling via gap junction channels (GJCs) and paracrine communication via the extracellular compartment, two routes to which channels composed of transmembrane connexin (Cx) or pannexin (Panx) proteins can contribute. Multiple isoforms of both protein families are present in the CNS and each CNS cell type is characterized by a unique Cx/Panx portfolio. Over the last two decades, research has uncovered a multilevel platform via which Cxs and Panxs can influence different cellular functions within a tissue: (1) Cx GJCs enable a direct cell-cell communication of small molecules, (2) Cx hemichannels and Panx channels can contribute to autocrine/paracrine signaling pathways, and (3) different structural domains of these proteins allow for channel-independent functions, such as cell-cell adhesion, interactions with the cytoskeleton, and the activation of intracellular signaling pathways. In this paper, we discuss current knowledge on their multifaceted contribution to brain development and to specific processes in the NGVU, including synaptic transmission and plasticity, glial signaling, vasomotor control, and blood-brain barrier integrity in the mature CNS. By highlighting both physiological and pathological conditions, it becomes evident that Cxs and Panxs can play a dual role in the CNS and that an accurate fine-tuning of each signaling mechanism is crucial for normal CNS physiology.
Collapse
Affiliation(s)
- Elke Decrock
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| | - Marijke De Bock
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| | - Nan Wang
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, KU Leuven, Louvain, Belgium
| | - Christian Giaume
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, 75231 Paris Cedex 05, France
- University Pierre et Marie
Curie, ED, N°158, 75005 Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, 75005 Paris, France
| | - Christian C. Naus
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Colin R. Green
- Department of Ophthalmology, The University of Auckland, Auckland, New Zealand
| | - Luc Leybaert
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| |
Collapse
|
49
|
Navarro G, Borroto-Escuela DO, Fuxe K, Franco R. Purinergic signaling in Parkinson's disease. Relevance for treatment. Neuropharmacology 2015. [PMID: 26211977 DOI: 10.1016/j.neuropharm.2015.07.024] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Purinergic signaling modulates dopaminergic neurotransmission in health and disease. Classically adenosine A1 and A2A receptors have been considered key for the fine tune control of dopamine actions in the striatum, the main CNS motor control center. The main adenosine signaling mechanism is via the cAMP pathway but the future will tell whether calcium signaling is relevant in adenosinergic control of striatal function. Very relevant is the recent approval in Japan of the adenosine A2A receptor antagonist, istradefylline, for use in Parkinson's disease patients. Purine nucleotides are also regulators of striatal dopamine neurotransmission via P2 purinergic receptors. In parallel to the alpha-synuclein hypothesis of Parkinson's disease etiology, purinergic P2X1 receptors have been identified as mediators of accumulation of the Lewy-body enriched protein alpha-synuclein. Of note is the expression in striatum of purinergic-receptor-containing heteromers that are potential targets of anti-Parkinson's disease therapies and should be taken into account in drug discovery programs. This article is part of the Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.
Collapse
Affiliation(s)
- Gemma Navarro
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| | - Dasiel O Borroto-Escuela
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Earth, Life and Environmental Sciences, Section of Physiology, Campus Scientifico Enrico Mattei, University of Urbino, Urbino, Italy.
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Rafael Franco
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
50
|
Bravo D, Maturana CJ, Pelissier T, Hernández A, Constandil L. Interactions of pannexin 1 with NMDA and P2X7 receptors in central nervous system pathologies: Possible role on chronic pain. Pharmacol Res 2015. [PMID: 26211949 DOI: 10.1016/j.phrs.2015.07.016] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Pannexin 1 (Panx1) is a glycoprotein that acts as a membrane channel in a wide variety of tissues in mammals. In the central nervous system (CNS) Panx1 is expressed in neurons, astrocytes and microglia, participating in the pathophysiology of some CNS diseases, such as epilepsy, anoxic depolarization after stroke and neuroinflammation. In these conditions Panx1 acts as an important modulator of the neuroinflammatory response, by secreting ATP, by interacting with the P2X7 receptor (P2X7R), and as an amplifier of NMDA receptor (NMDAR) currents, particularly in conditions of pathological neuronal hyperexcitability. Here, we briefly reviewed the current evidences that support the interaction of Panx1 with NMDAR and P2X7R in pathological contexts of the CNS, with special focus in recent data supporting that Panx1 is involved in chronic pain signaling by interacting with NMDAR in neurons and with P2X7R in glia. The participation of Panx1 in chronic pain constitutes a novel topic for research in the field of clinical neurosciences and a potential target for pharmacological interventions in chronic pain.
Collapse
Affiliation(s)
- D Bravo
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Chile; School of Kinesiology, Faculty of Sport, Health and Recreation, University Bernardo O'Higgins, Chile.
| | - C J Maturana
- Departamento de Fisiología, Pontificia Universidad Católica De Chile, Chile
| | - T Pelissier
- Program of Molecular and Clinical Pharmacology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Chile
| | - A Hernández
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Chile
| | - L Constandil
- Laboratory of Neurobiology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Chile
| |
Collapse
|