Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
Neuroscience ; 285: 166-93, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25451276

ABSTRACT

Gap junctions provide for direct intercellular electrical and metabolic coupling. The abundance of gap junctions at "large myelinated club ending (LMCE)" synapses on Mauthner cells (M-cells) of the teleost brain provided a convenient model to correlate anatomical and physiological properties of electrical synapses. There, presynaptic action potentials were found to evoke short-latency electrical "pre-potentials" immediately preceding their accompanying glutamate-induced depolarizations, making these the first unambiguously identified "mixed" (i.e., chemical plus electrical) synapses in the vertebrate CNS. We recently showed that gap junctions at these synapses exhibit asymmetric electrical resistance (i.e., electrical rectification), which we correlated with total molecular asymmetry of connexin composition in their apposing gap junction hemiplaques, with connexin35 (Cx35) restricted to axon terminal hemiplaques and connexin34.7 (Cx34.7) restricted to apposing M-cell plasma membranes. We now show that similarly heterotypic neuronal gap junctions are abundant throughout goldfish brain, with labeling exclusively for Cx35 in presynaptic hemiplaques and exclusively for Cx34.7 in postsynaptic hemiplaques. Moreover, the vast majority of these asymmetric gap junctions occur at glutamatergic axon terminals. The widespread distribution of heterotypic gap junctions at glutamatergic mixed synapses throughout goldfish brain and spinal cord implies that pre- vs. postsynaptic asymmetry at electrical synapses evolved early in the chordate lineage. We propose that the advantages of the molecular and functional asymmetry of connexins at electrical synapses that are so prominently expressed in the teleost CNS are unlikely to have been abandoned in higher vertebrates. However, to create asymmetric coupling in mammals, where most gap junctions are composed of connexin36 (Cx36) on both sides, would require some other mechanism, such as differential phosphorylation of connexins on opposite sides of the same gap junction or on asymmetric differences in the complement of their scaffolding and regulatory proteins.


Subject(s)
Brain/metabolism , Gap Junctions/metabolism , Glutamic Acid/metabolism , Goldfish/metabolism , Synapses/metabolism , Animals , Axons/metabolism , Dendrites/metabolism , Fish Proteins/metabolism , Immunohistochemistry , Microscopy, Confocal , Microscopy, Electron
2.
Neuroscience ; 149(2): 350-71, 2007 Oct 26.
Article in English | MEDLINE | ID: mdl-17904757

ABSTRACT

Suprachiasmatic nucleus (SCN) neurons generate circadian rhythms, and these neurons normally exhibit loosely-synchronized action potentials. Although electrotonic coupling has long been proposed to mediate this neuronal synchrony, ultrastructural studies have failed to detect gap junctions between SCN neurons. Nevertheless, it has been proposed that neuronal gap junctions exist in the SCN; that they consist of connexin32 or, alternatively, connexin36; and that connexin36 knockout eliminates neuronal coupling between SCN neurons and disrupts circadian rhythms. We used confocal immunofluorescence microscopy and freeze-fracture replica immunogold labeling to examine the distributions of connexin30, connexin32, connexin36, and connexin43 in rat and mouse SCN and used whole-cell recordings to re-assess electrotonic and tracer coupling. Connexin32-immunofluorescent puncta were essentially absent in SCN but connexin36 was relatively abundant. Fifteen neuronal gap junctions were identified ultrastructurally, all of which contained connexin36 but not connexin32, whereas nearby oligodendrocyte gap junctions contained connexin32. In adult SCN, one neuronal gap junction was >600 connexons, whereas 75% were smaller than 50 connexons, which may be below the limit of detectability by fluorescence microscopy and thin-section electron microscopy. Whole-cell recordings in hypothalamic slices revealed tracer coupling with neurobiotin in <5% of SCN neurons, and paired recordings (>40 pairs) did not reveal obvious electrotonic coupling or synchronized action potentials, consistent with few neurons possessing large gap junctions. However, most neurons had partial spikes or spikelets (often <1 mV), which remained after QX-314 [N-(2,6-dimethylphenylcarbamoylmethyl)triethylammonium bromide] had blocked sodium-mediated action potentials within the recorded neuron, consistent with spikelet transmission via small gap junctions. Thus, a few "miniature" gap junctions on most SCN neurons appear to mediate weak electrotonic coupling between limited numbers of neuron pairs, thus accounting for frequent detection of partial spikes and hypothetically providing the basis for "loose" electrical or metabolic synchronization of electrical activity commonly observed in SCN neuronal populations during circadian rhythms.


Subject(s)
Connexins/physiology , Gap Junctions/physiology , Suprachiasmatic Nucleus/physiology , Animals , Connexins/genetics , Detergents/pharmacology , Electrophysiology , Freeze Fracturing , Immunohistochemistry , Male , Mice , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron , Neuroglia/physiology , Neurons/physiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Sodium Dodecyl Sulfate/pharmacology , Gap Junction beta-1 Protein , Gap Junction delta-2 Protein
3.
Neuroscience ; 147(4): 938-56, 2007 Jul 29.
Article in English | MEDLINE | ID: mdl-17601673

ABSTRACT

Locus coeruleus neurons are strongly coupled during early postnatal development, and it has been proposed that these neurons are linked by extraordinarily abundant gap junctions consisting of connexin32 (Cx32) and connexin26 (Cx26), and that those same connexins abundantly link neurons to astrocytes. Based on the controversial nature of those claims, immunofluorescence imaging and freeze-fracture replica immunogold labeling were used to re-investigate the abundance and connexin composition of neuronal and glial gap junctions in developing and adult rat and mouse locus coeruleus. In early postnatal development, connexin36 (Cx36) and connexin43 (Cx43) immunofluorescent puncta were densely distributed in the locus coeruleus, whereas Cx32 and Cx26 were not detected. By freeze-fracture replica immunogold labeling, Cx36 was found in ultrastructurally-defined neuronal gap junctions, whereas Cx32 and Cx26 were not detected in neurons and only rarely detected in glia. In 28-day postnatal (adult) rat locus coeruleus, immunofluorescence labeling for Cx26 was always co-localized with the glial gap junction marker Cx43; Cx32 was associated with the oligodendrocyte marker 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNPase); and Cx36 was never co-localized with Cx26, Cx32 or Cx43. Ultrastructurally, Cx36 was localized to gap junctions between neurons, whereas Cx32 was detected only in oligodendrocyte gap junctions; and Cx26 was found only rarely in astrocyte junctions but abundantly in pia mater. Thus, in developing and adult locus coeruleus, neuronal gap junctions contain Cx36 but do not contain detectable Cx32 or Cx26, suggesting that the locus coeruleus has the same cell-type specificity of connexin expression as observed ultrastructurally in other regions of the CNS. Moreover, in both developing and adult locus coeruleus, no evidence was found for gap junctions or connexins linking neurons with astrocytes or oligodendrocytes, indicating that neurons in this nucleus are not linked to the pan-glial syncytium by Cx32- or Cx26-containing gap junctions or by abundant free connexons composed of those connexins.


Subject(s)
Connexins/metabolism , Gap Junctions/metabolism , Locus Coeruleus/cytology , Neurons/cytology , Rodentia/metabolism , Animals , Animals, Newborn , Connexins/classification , Connexins/deficiency , Freeze Fracturing/methods , Gap Junctions/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Scanning/methods , Nerve Tissue Proteins/metabolism , Rats , Rats, Sprague-Dawley , Rodentia/growth & development , Gap Junction delta-2 Protein
4.
Neuroscience ; 142(4): 1093-117, 2006 Nov 03.
Article in English | MEDLINE | ID: mdl-17010526

ABSTRACT

Neuronal gap junctions are abundant in both outer and inner plexiform layers of the mammalian retina. In the inner plexiform layer (IPL), ultrastructurally-identified gap junctions were reported primarily in the functionally-defined and anatomically-distinct ON sublamina, with few reported in the OFF sublamina. We used freeze-fracture replica immunogold labeling and confocal microscopy to quantitatively analyze the morphologies and distributions of neuronal gap junctions in the IPL of adult rat and mouse retina. Under "baseline" conditions (photopic illumination/general anesthesia), 649 neuronal gap junctions immunogold-labeled for connexin36 were identified in rat IPL, of which 375 were photomapped to OFF vs. ON sublaminae. In contrast to previous reports, the volume-density of gap junctions was equally abundant in both sublaminae. Five distinctive morphologies of gap junctions were identified: conventional crystalline and non-crystalline "plaques" (71% and 3%), plus unusual "string" (14%), "ribbon" (7%) and "reticular" (2%) forms. Plaque and reticular gap junctions were distributed throughout the IPL. However, string and ribbon gap junctions were restricted to the OFF sublamina, where they represented 48% of gap junctions in that layer. In string and ribbon junctions, curvilinear strands of connexons were dispersed over 5 to 20 times the area of conventional plaques having equal numbers of connexons. To define morphologies of gap junctions under different light-adaptation conditions, we examined an additional 1150 gap junctions from rats and mice prepared after 30 min of photopic, mesopic and scotopic illumination, with and without general anesthesia. Under these conditions, string and ribbon gap junctions remained abundant in the OFF sublamina and absent in the ON sublamina. Abundant gap junctions in the OFF sublamina of these two rodents with rod-dominant retinas revealed previously-undescribed but extensive pathways for inter-neuronal communication; and the wide dispersion of connexons in string and ribbon gap junctions suggests unique structural features of gap junctional coupling in the OFF vs. ON sublamina.


Subject(s)
Gap Junctions/ultrastructure , Neural Pathways/ultrastructure , Neurons/ultrastructure , Retina/ultrastructure , Animals , Cell Communication/physiology , Cell Membrane/physiology , Cell Membrane/ultrastructure , Female , Freeze Fracturing , Gap Junctions/physiology , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Immunoelectron , Models, Neurological , Neural Pathways/physiology , Neurons/physiology , Photic Stimulation , Rats , Rats, Sprague-Dawley , Retina/physiology , Retinal Rod Photoreceptor Cells/physiology , Retinal Rod Photoreceptor Cells/ultrastructure , Vision, Ocular/physiology
5.
Neuroscience ; 140(2): 433-51, 2006 Jun 30.
Article in English | MEDLINE | ID: mdl-16650609

ABSTRACT

Most gap junctions between neurons in mammalian retina contain abundant connexin36, often in association with the scaffolding protein zonula occludens-1. We now investigate co-association of connexin36, zonula occludens-1, zonula occludens-2 and Y-box transcription factor 3 (zonula occludens-1-associated nucleic acid-binding protein) in mouse and rat retina. By immunoblotting, zonula occludens-1-associated nucleic acid-binding protein and zonula occludens-2 were both detected in retina, and zonula occludens-2 in retina was found to co-immunoprecipitate with connexin36. By immunofluorescence, the four proteins appeared as puncta distributed in the plexiform layers. In the inner plexiform layer, most connexin36-puncta were co-localized with zonula occludens-1, and many were co-localized with zonula occludens-1-associated nucleic acid-binding protein. Moreover, zonula occludens-1-associated nucleic acid-binding protein was often co-localized with zonula occludens-1. Nearly all zonula occludens-2-puncta were positive for connexin36, zonula occludens-1 and zonula occludens-1-associated nucleic acid-binding protein. In the outer plexiform layer, connexin36 was also often co-localized with zonula occludens-1-associated nucleic acid-binding protein. In connexin36 knockout mice, labeling of zonula occludens-1 was slightly reduced in the inner plexiform layer, zonula occludens-1-associated nucleic acid-binding protein was decreased in the outer plexiform layer, and both zonula occludens-1-associated nucleic acid-binding protein and zonula occludens-2 were markedly decreased in the inner sublamina of the inner plexiform layer, whereas zonula occludens-1, zonula occludens-2 and zonula occludens-1-associated nucleic acid-binding protein puncta persisted and remained co-localized in the outer sublamina of the inner plexiform layer. By freeze-fracture replica immunogold labeling, connexin36 was found to be co-localized with zonula occludens-2 within individual neuronal gap junctions. In addition, zonula occludens-1-associated nucleic acid-binding protein was abundant in a portion of ultrastructurally-defined gap junctions throughout the inner plexiform layer, and some of these junctions contained both connexin36 and zonula occludens-1-associated nucleic acid-binding protein. These distinct patterns of connexin36 association with zonula occludens-1, zonula occludens-2 and zonula occludens-1-associated nucleic acid-binding protein in different sublaminae of retina, and differential responses of these proteins to connexin36 gene deletion suggest differential regulatory and scaffolding roles of these gap junction accessory proteins. Further, the persistence of a subpopulation of zonula occludens-1/zonula occludens-2/zonula occludens-1-associated nucleic acid-binding protein co-localized puncta in the outer part of the inner plexiform layer of connexin36 knockout mice suggests close association of these proteins with other structures in retina, possibly including gap junctions composed of an as-yet-unidentified connexin.


Subject(s)
Connexins/metabolism , DNA-Binding Proteins/metabolism , Gap Junctions/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Retina/metabolism , Animals , Calbindin 2 , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Connexins/genetics , DNA-Binding Proteins/genetics , Female , Fluorescent Antibody Technique , Freeze Fracturing , Gap Junctions/ultrastructure , Immunohistochemistry , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Scanning , Neurons/metabolism , Neurons/ultrastructure , Phosphoproteins/genetics , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , RNA-Binding Proteins/genetics , Rats , Rats, Sprague-Dawley , Retina/ultrastructure , S100 Calcium Binding Protein G/metabolism , Zonula Occludens-1 Protein , Zonula Occludens-2 Protein , Gap Junction delta-2 Protein
6.
Neuroscience ; 136(1): 65-86, 2005.
Article in English | MEDLINE | ID: mdl-16203097

ABSTRACT

The subcellular distributions and co-associations of the gap junction-forming proteins connexin 47 and connexin 32 were investigated in oligodendrocytes of adult mouse and rat CNS. By confocal immunofluorescence light microscopy, abundant connexin 47 was co-localized with astrocytic connexin 43 on oligodendrocyte somata, and along myelinated fibers, whereas connexin 32 without connexin 47 was co-localized with contactin-associated protein (caspr) in paranodes. By thin-section transmission electron microscopy, connexin 47 immunolabeling was on the oligodendrocyte side of gap junctions between oligodendrocyte somata and astrocytes. By freeze-fracture replica immunogold labeling, large gap junctions between oligodendrocyte somata and astrocyte processes contained much more connexin 47 than connexin 32. Along surfaces of internodal myelin, connexin 47 was several times as abundant as connexin 32, and in the smallest gap junctions, often occurred without connexin 32. In contrast, connexin 32 was localized without connexin 47 in newly-described autologous gap junctions in Schmidt-Lanterman incisures and between paranodal loops bordering nodes of Ranvier. Thus, connexin 47 in adult rodent CNS is the most abundant connexin in most heterologous oligodendrocyte-to-astrocyte gap junctions, whereas connexin 32 is the predominant if not sole connexin in autologous ("reflexive") oligodendrocyte gap junctions. These results clarify the locations and connexin compositions of heterologous and autologous oligodendrocyte gap junctions, identify autologous gap junctions at paranodes as potential sites for modulating paranodal electrical properties, and reveal connexin 47-containing and connexin 32-containing gap junctions as conduits for long-distance intracellular and intercellular movement of ions and associated osmotic water. The autologous gap junctions may regulate paranodal electrical properties during saltatory conduction. Acting in series and in parallel, autologous and heterologous oligodendrocyte gap junctions provide essential pathways for intra- and intercellular ionic homeostasis.


Subject(s)
Central Nervous System/metabolism , Connexins/metabolism , Gap Junctions/metabolism , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Central Nervous System/cytology , Central Nervous System/ultrastructure , Connexin 43/metabolism , Cytoplasm/metabolism , Female , Fluorescent Antibody Technique , Freeze Fracturing , Gap Junctions/ultrastructure , Homeostasis , Immunohistochemistry , Ions , Male , Mice , Mice, Inbred Strains , Microscopy, Electron , Myelin Sheath/ultrastructure , Oligodendroglia/ultrastructure , Potassium/metabolism , Ranvier's Nodes/metabolism , Rats , Rats, Sprague-Dawley , Subcellular Fractions/metabolism , Tissue Distribution , Gap Junction beta-1 Protein
7.
Neuroscience ; 126(3): 611-30, 2004.
Article in English | MEDLINE | ID: mdl-15183511

ABSTRACT

Gap junctions between glial cells in mammalian CNS are known to contain several connexins (Cx), including Cx26, Cx30 and Cx43 at astrocyte-to-astrocyte junctions, and Cx29 and Cx32 on the oligodendrocyte side of astrocyte-to-oligodendrocyte junctions. Recent reports indicating that oligodendrocytes also express Cx47 prompted the present studies of Cx47 localization and relationships to other glial connexins in mouse CNS. In view of the increasing number of connexins reported to interact directly with the scaffolding protein zonula occludens-1 (ZO-1), we investigated ZO-1 expression and Cx47/ZO-1 interaction capabilities in brain, spinal cord and Cx47-transfected HeLa cells. From counts of over 9000 oligodendrocytes labeled by immunofluorescence in various brain regions, virtually all of these cells were found to express Cx29, Cx32 and Cx47. Oligodendrocyte somata displayed robust Cx47-immunopositive puncta that were co-localized with punctate labeling for Cx32 and Cx43. By freeze-fracture replica immunogold labeling, Cx47 was abundant on the oligodendrocyte-side of oligodendrocyte/astrocyte gap junctions. By immunofluorescence, labeling for Cx47 along myelinated fibers was sparse in most brain regions, whereas Cx29 and Cx32 were previously found to be concentrated along these fibers. By immunogold labeling, Cx47 was found in numerous small gap junctions linking myelin to astrocytes, but not within deeper layers of myelin. Brain subcellular fractionation revealed a lack of Cx47 enrichment in myelin fractions, which nevertheless contained an enrichment of Cx32 and Cx29. Oligodendrocytes were immunopositive for ZO-1, and displayed almost total Cx47/ZO-1 co-localization. ZO-1 was found to co-immunoprecipitate with Cx47, and pull-down assays indicated binding of Cx47 to the second PDZ domain of ZO-1. Our results indicate widespread expression of Cx47 by oligodendrocytes, but with a distribution pattern in relative levels inverse to the abundance of Cx29 in myelin and paucity of Cx29 in oligodendrocyte somata. Further, our findings suggest a scaffolding and/or regulatory role of ZO-1 at the oligodendrocyte side of astrocyte-to-oligodendrocyte gap junctions.


Subject(s)
Cell Communication/physiology , Connexins/biosynthesis , Oligodendroglia/metabolism , Tight Junctions/metabolism , Animals , Astrocytes/metabolism , Astrocytes/ultrastructure , Blotting, Western , Brain/metabolism , Connexin 26 , Fluorescent Antibody Technique , Freeze Fracturing , Gap Junctions/metabolism , Gap Junctions/ultrastructure , HeLa Cells , Humans , Male , Membrane Proteins/metabolism , Mice , Microscopy, Confocal , Oligodendroglia/ultrastructure , Phosphoproteins/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/metabolism , Tight Junctions/ultrastructure , Zonula Occludens-1 Protein
8.
J Neurocytol ; 33(1): 131-51, 2004 Jan.
Article in English | MEDLINE | ID: mdl-15173637

ABSTRACT

Combined confocal microscopy and freeze-fracture replica immunogold labeling (FRIL) were used to examine the connexin identity at electrical synapses in goldfish brain and rat retina, and to test for "co-localization" vs. "close proximity" of connexins to other functionally interacting proteins in synapses of goldfish and mouse brain and rat retina. In goldfish brain, confocal microscopy revealed immunofluorescence for connexin35 (Cx35) and NMDA-R1 (NR1) glutamate receptor protein in Mauthner Cell/Club Ending synapses. By FRIL double labeling, NR1 glutamate receptors were found in clusters of intramembrane particles in the postsynaptic membrane extraplasmic leaflets, and these distinctive postsynaptic densities were in close proximity (0.1-0.3 microm) to neuronal gap junctions labeled for Cx35, which is the fish ortholog of connexin36 (Cx36) found at neuronal gap junctions in mammals. Immunogold labeling for Cx36 in adult rat retina revealed abundant gap junctions, including several previously unrecognized morphological types. As in goldfish hindbrain, immunogold double labeling revealed NR1-containing postsynaptic densities localized near Cx36-labeled gap junction in rat inferior olive. Confocal immunofluorescence microscopy revealed widespread co-localization of Cx36 and ZO-1, particularly in the reticular thalamic nucleus and amygdala of mouse brain. By FRIL, ZO-1 immunoreactivity was co-localized with Cx36 at individual gap junction plaques in rat retinal neurons. As cytoplasmic accessory proteins, ZO-1 and possibly related members of the membrane-associated guanylate kinase (MAGUK) family represent scaffolding proteins that may bind to and regulate the activity of many neuronal gap junctions. These data document the power of combining immunofluorescence confocal microscopy with FRIL ultrastructural imaging and immunogold labeling to determine the relative proximities of proteins that are involved in short- vs. intermediate-range molecular interactions in the complex membrane appositions at synapses between neurons.


Subject(s)
Brain Mapping/methods , Connexins/analysis , Eye Proteins/analysis , Membrane Proteins/analysis , Phosphoproteins/analysis , Proteomics/methods , Receptors, N-Methyl-D-Aspartate/analysis , Animals , Connexins/biosynthesis , Eye Proteins/biosynthesis , Goldfish , Immunohistochemistry , Membrane Proteins/biosynthesis , Mice , Mice, Knockout , Phosphoproteins/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/biosynthesis , Zonula Occludens-1 Protein , Gap Junction delta-2 Protein
9.
Cell Commun Adhes ; 10(4-6): 419-23, 2003.
Article in English | MEDLINE | ID: mdl-14681051

ABSTRACT

Auditory afferents terminating as mixed, electrical, and chemical, synapses on the goldfish Mauthner cells constitute an ideal experimental model to study the properties of gap junctions in the nervous system as well as to explore possible functional interactions with the other major form of interneuronal communication--chemically mediated synapses. By combining confocal microscopy and freeze-fracture replica immunogold labeling (FRIL), we found that gap junctions at these synapses contain connexin35 (Cx35), the fish ortholog of the neuron-specific human and mouse connexin36 (Cx36). Conductance of gap junction channels at these endings is known to be dynamically modulated by the activity of their co-localized chemically mediated glutamatergic synapses. By using simultaneous pre- and postsynaptic recordings at these single terminals, we demonstrate that such functional interaction takes place in the same ending, within a few micrometers. Accordingly, we also found evidence by confocal and FRIL double-immunogold labeling that the NR1 subunit of the NMDA glutamate receptor, proposed to be a key regulatory element, is present at postsynaptic densities closely associated with gap junction plaques containing Cx35. Given the widespread distribution of Cx35- and Cx36-mediated electrical synapses and glutamatergic synapses, our data suggest that the local functional interactions observed at these identifiable junctions may also apply to other electrical synapses, including those in mammalian brain.


Subject(s)
Connexins/physiology , Eye Proteins/physiology , Gap Junctions/physiology , Goldfish/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission/physiology , Animals , Connexins/genetics , Electric Conductivity , Excitatory Postsynaptic Potentials/physiology , Eye Proteins/genetics , Freeze Fracturing , Presynaptic Terminals/physiology , Gap Junction delta-2 Protein
10.
J Neurosci ; 23(20): 7489-503, 2003 Aug 20.
Article in English | MEDLINE | ID: mdl-12930787

ABSTRACT

Auditory afferents terminating as "large myelinated club endings" on goldfish Mauthner cells are identifiable "mixed" (electrical and chemical) synaptic terminals that offer the unique opportunity to correlate physiological properties with biochemical composition and specific ultrastructural features of individual synapses. By combining confocal microscopy and freeze-fracture replica immunogold labeling (FRIL), we demonstrate that gap junctions at these synapses contain connexin35 (Cx35). This connexin is the fish ortholog of the neuron-specific human and mouse connexin36 that is reported to be widely distributed in mammalian brain and to be responsible for electrical coupling between many types of neurons. Similarly, connexin35 was found at gap junctions between neurons in other brain regions, suggesting that connexin35-mediated electrical transmission is common in goldfish brain. Conductance of gap junction channels at large myelinated club endings is known to be dynamically modulated by the activity of their colocalized glutamatergic synapses. We show evidence by confocal microscopy for the presence of the NR1 subunit of the NMDA glutamate receptor subtype, proposed to be a key regulatory element, at these large endings. Furthermore, we also show evidence by FRIL double-immunogold labeling that the NR1 subunit of the NMDA glutamate receptor is present at postsynaptic densities closely associated with gap junction plaques containing Cx35 at mixed synapses across the goldfish hindbrain. Given the widespread distribution of electrical synapses and glutamate receptors, our results suggest that the plastic properties observed at these identifiable junctions may apply to other electrical synapses, including those in mammalian brain.


Subject(s)
Connexins/physiology , Neurons/physiology , Synapses/physiology , Synaptic Transmission , Animals , Antibody Specificity , Astrocytes/chemistry , Astrocytes/ultrastructure , Auditory Pathways , Central Nervous System/physiology , Connexins/analysis , Connexins/immunology , Electric Conductivity , Eye Proteins/physiology , Gap Junctions/chemistry , Gap Junctions/ultrastructure , Goldfish , Immunohistochemistry , Microscopy, Confocal , Nerve Endings/chemistry , Nerve Endings/cytology , Neuronal Plasticity , Neurons/chemistry , Neurons/cytology , Presynaptic Terminals/chemistry , Receptors, N-Methyl-D-Aspartate/analysis , Rhombencephalon/physiology , Synapses/chemistry , Synapses/ultrastructure , Gap Junction delta-2 Protein
11.
Appl Microbiol Biotechnol ; 55(4): 471-5, 2001 May.
Article in English | MEDLINE | ID: mdl-11398929

ABSTRACT

The number of foreign protein molecules expressed on the cell surface of the budding yeast Saccharomyces cerevisiae by cell surface engineering was quantitatively evaluated using enhanced green fluorescent protein (EGFP). The emission from EGFP on the cell surface was affected by changes in pH. The amount of EGFP on the cell surface, displayed as alpha-agglutinin-fusion protein under control of the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) promoter, was determined at the optimum pH of 7.0. The fluorometric analysis and the image analysis by confocal laser scanning microscopy (CLSM) showed a similar number of molecules displayed on the cell surface, demonstrating that 10(4)-10(5) molecules of alpha-agglutinin-fused molecules per cell were expressed. Furthermore, the amount of fluorescent protein expressed on cells harboring a multicopy plasmid was three to four times higher than that on cells harboring the gene integrated into the genome.


Subject(s)
Arabidopsis/growth & development , Saccharomyces cerevisiae/genetics , Base Sequence , Cell Membrane/metabolism , DNA Primers , Evaluation Studies as Topic , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Hydrogen-Ion Concentration , Microscopy, Confocal , Microscopy, Fluorescence , Promoter Regions, Genetic
12.
Yeast ; 18(5): 433-44, 2001 Mar 30.
Article in English | MEDLINE | ID: mdl-11255251

ABSTRACT

The chemical composition of the cell wall of Sz. pombe is known as beta-1,3-glucan, beta-1,6-glucan, alpha-1,3-glucan and alpha-galactomannan; however, the three-dimensional interactions of those macromolecules have not yet been clarified. Transmission electron microscopy reveals a three-layered structure: the outer layer is electron-dense, the adjacent layer is less dense, and the third layer bordering the cell membrane is dense. In intact cells of Sz. pombe, the high-resolution scanning electron microscope reveals a surface completely filled with alpha-galactomannan particles. To better understand the organization of the cell wall and to complement our previous studies, we set out to locate the three different types of beta-glucan by immuno-electron microscopy. Our results suggest that the less dense layer of the cell wall contains mainly beta-1,6-branched beta-1,3-glucan. Occasionally a line of gold particles can be seen, labelling fine filaments radiating from the cell membrane to the alpha-galactomannan layer, suggesting that some of the radial filaments contain beta-1,6-branched beta-1,3-glucan. beta-1,6-glucan is preferentially located underneath the alpha-galactomannan layer. Linear beta-1,3-glucan is exclusively located in the primary septum of dividing cells. beta-1,6-glucan only labels the secondary septum and does not co-localize with linear beta-1,3-glucan, while beta-1,6-branched beta-1,3-glucan is present in both septa. Linear beta-1,3-glucan is present from early stages of septum formation and persists until the septum is completely formed; then just before cell division the label disappears. From these results we suggest that linear beta-1,3-glucan is involved in septum formation and perhaps the separation of the two daughter cells. In addition, we frequently found beta-1,6-glucan label on the Golgi apparatus, on small vesicles and underneath the cell membrane. These results give fresh evidence for the hypothesis that beta-1,6-glucan is synthesized in the endoplasmic reticulum-Golgi system and exported to the cell membrane.


Subject(s)
Cell Wall/ultrastructure , Glucans/metabolism , Microscopy, Immunoelectron/methods , Schizosaccharomyces/ultrastructure , Cell Wall/metabolism , Golgi Apparatus/metabolism , Schizosaccharomyces/metabolism
13.
Appl Microbiol Biotechnol ; 57(4): 528-33, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11762600

ABSTRACT

We have successfully created a novel yeast strain able to monitor changes in environmental conditions by displaying either green fluorescent protein (GFP) from Aequorea victoria or blue fluorescent protein (BFP), a variant of GFP, on its cell surface as a visible reporter. For the display of these fluorescent proteins on the cell surface of Saccharomyces cerevisiase, our cell-surface-engineering system was utilized. The GAPDH promoter, which is active in the presence of glucose, and the UPR-ICL promoter from Candida tropicalis, which starts to function in the presence of a reduced level of glucose, were employed simultaneously to express the GFP-encoding gene and the BFP-encoding gene, respectively. This cell-surface-engineered yeast strain emitted green fluorescence from the cell surface when sufficient glucose was present in the medium, and blue fluorescence from the same cell surface when the glucose in the medium was consumed. The fluorescent proteins displayed on the cell surface using the different promoters enabled us to monitor the concentrations of intra- and/or extracellular glucose that regulated activation or inactivation of the promoters. This novel yeast strain could facilitate the computerized control of various bioprocesses measuring emitted fluorescence.


Subject(s)
Genetic Engineering , Glucose/pharmacology , Luminescent Proteins/biosynthesis , Saccharomyces cerevisiae/genetics , Green Fluorescent Proteins , Promoter Regions, Genetic
14.
FEMS Microbiol Lett ; 192(2): 243-8, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11064202

ABSTRACT

A starch-utilizing Saccharomyces cerevisiae strain was constructed by cell surface engineering. Distribution of the heterologous glucoamylase-alpha-agglutinin fusion protein on the yeast cell was analyzed by indirect fluorescence microscopy using an anti-glucoamylase antibody. Most of the intense fluorescence was first localized in the small bud, then observed on the entire cell wall of the daughter and mother cells. Fluorescence also accumulated at the neck region. These observations suggest that the display of the heterologous protein on the cell surface is carried with other cell wall components to the areas in which the cell wall is newly synthesized; the distribution is controlled by the cell cycle. Then, the heterologous protein-encoding gene was expressed in a sec1 mutant, in which secretory vesicles accumulate under restrictive temperature, and the produced protein was detected by immunoelectron microscopy. Most of the gold particles that reacted with the fusion protein were not localized in vesicles but in expanding endoplasmic reticulum. This phenomenon may be due to overproduction of the heterologous protein which was designed to be displayed on the cell wall. Artificial production of heterologous protein may have caused a relative shortage of glycosyl phosphatidylinositol anchors.


Subject(s)
Cell Wall/enzymology , Glucan 1,4-alpha-Glucosidase/analysis , Saccharomyces cerevisiae/enzymology , Agglutinins/genetics , Cell Cycle , Cell Wall/genetics , Fluorescent Antibody Technique, Indirect , Fungal Proteins/analysis , Fungal Proteins/genetics , Glucan 1,4-alpha-Glucosidase/genetics , Microscopy, Immunoelectron , Mutation , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/genetics , Saccharomyces cerevisiae/genetics , Starch/metabolism
15.
Appl Microbiol Biotechnol ; 54(1): 90-6, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10952010

ABSTRACT

An engineered yeast with emission of fluorescence from the cell surface was constructed. Cell surface engineering was applied to display a visible reporter molecule, green fluorescent protein (GFP). A glucose-inducible promoter GAPDH as a model promoter was selected to control the expression of the reporter gene in response to environmental changes. The GFP gene was fused with the gene encoding the C-terminal half of alpha-agglutinin of Saccharomyces cerevisiae having a glycosylphosphatidylinositol anchor attachment signal sequence. A secretion signal sequence of the fungal glucoamylase precursor protein was connected to the N-terminal of GFP. This designed gene was integrated into the TRP1 locus of the chromosome of S. cerevisiae with homologous recombination. Fluorescence microscopy demonstrated that the transformant cells emitted green fluorescence derived from functionally expressed GFP involved in the fusion molecule. The surface display of GFP was further verified by immunofluorescence labeling with a polyclonal antibody (raised in rabbits) against GFP as the first antibody and Rhodamine Red-X-conjugated goat anti-rabbit IgG as the second antibody which cannot penetrate into the cell membrane. The display of GFP on the cell surface was confirmed using a confocal laser scanning microscope and by measuring fluorescence in each cell fraction obtained after the subcellular fractionation. As GFP was proved to be displayed as an active form on the cell surface, selection of promoters will endow yeast cells with abilities to respond to changes in environmental conditions, including nutrient concentrations in the media, through the emission of fluorescence.


Subject(s)
Glucose/metabolism , Luminescent Proteins/genetics , Recombinant Fusion Proteins/genetics , Saccharomyces cerevisiae/genetics , Base Sequence , Cell Membrane/metabolism , DNA Primers , Electrophoresis, Polyacrylamide Gel , Fluorescence , Green Fluorescent Proteins , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods
16.
Planta ; 211(2): 265-74, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10945221

ABSTRACT

NADP-dependent malic enzyme (NADP-ME) is a major decarboxylating enzyme in NADP-ME-type C4 species such as maize and Flaveria. In this study, chloroplastic NADP-ME was transferred to rice (Oryza sativa L.) using a chimeric gene composed of maize NADP-ME cDNA under the control of rice light-harvesting chlorophyll-a/b-binding protein (Cab) promoter. There was a 20- to 70-fold increase in the NADP-ME activity in leaves of transgenic rice compared to that in wild-type rice plants. Immunocytochemical studies by electron microscopy showed that maize NADP-ME was mostly localized in chloroplasts in transgenic rice plants, and that the chloroplasts were agranal without thylakoid stacking. Chlorophyll content and photosystem II activity were inversely correlated with the level of NADP-ME activity. These results suggest that aberrant chloroplasts in transgenic plants may be caused by excessive NADP-ME activity. Based on these results and the known fact that only bundle sheath cells of NADP-ME species, among all three C4 subgroups, have agranal chloroplasts, we postulate that a high level of chloroplastic NADP-ME activity could strongly affect the development of chloroplasts.


Subject(s)
Chloroplasts/metabolism , Malate Dehydrogenase/genetics , Oryza/physiology , Plants, Genetically Modified/physiology , Zea mays/enzymology , Chlorophyll/metabolism , Chlorophyll A , Chloroplasts/genetics , Chloroplasts/ultrastructure , Light-Harvesting Protein Complexes , Malate Dehydrogenase/metabolism , Oryza/enzymology , Oryza/genetics , Phenotype , Photosynthetic Reaction Center Complex Proteins/genetics , Photosystem II Protein Complex , Plant Leaves/physiology , Plant Leaves/ultrastructure , Promoter Regions, Genetic , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Zea mays/genetics
17.
J Electron Microsc (Tokyo) ; 48(2): 173-6, 1999.
Article in English | MEDLINE | ID: mdl-10356790

ABSTRACT

Modified genes of peroxisomal isocitrate lyase of Candida tropicalis (CT-ICL) were constructed and expressed in Saccharomyces cerevisiae cells. We observed subcellular localization of expressed products of the mutant CT-ICL genes by immunoelectron microscopy. An unknown structure termed a protein aggregate body (PAB) storing the expressed product was observed in cytoplasm in various mutants (Kamasawa et al. (1996) J. Electron Microsc. 45: 491-497). We chose two typical cells harbouring the mutant ICL genes delta 550 and delta 237-339 to analyse the ultrastructure and three-dimensional (3D) structure of PABs. The PABs had a homogeneous matrix with a wavy periphery in the cell image using a high-pressure freezing fixation method. Although PABs could not be separated from the cytoplasm or mitochondria under a confocal fluorescence microscope, 3D reconstruction of serial electron micrographs clearly showed the PAB was an independent structure of varying size and had the shape of an incomplete sphere. A cell was sometimes observed to have multiple PABs.


Subject(s)
Inclusion Bodies/ultrastructure , Isocitrate Lyase/analysis , Saccharomyces cerevisiae/enzymology , Candida/enzymology , Candida/genetics , Freezing , Image Processing, Computer-Assisted , Inclusion Bodies/enzymology , Isocitrate Lyase/genetics , Microscopy, Confocal , Microscopy, Immunoelectron , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/ultrastructure
18.
Appl Microbiol Biotechnol ; 51(1): 65-70, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10077821

ABSTRACT

The construction of a whole-cell biocatalyst with its sequential reaction has been performed by the genetic immobilization of two amylolytic enzymes on the yeast cell surface. A recombinant strain of Saccharomyces cerevisiae that displays glucoamylase and alpha-amylase on its cell surface was constructed and its starch-utilizing ability was evaluated. The gene encoding Rhizopus oryzae glucoamylase, with its own secretion signal peptide, and a truncated fragment of the alpha-amylase gene from Bacillus stearothermophilus with the prepro secretion signal sequence of the yeast alpha factor, respectively, were fused with the gene encoding the C-terminal half of the yeast alpha-agglutinin. The constructed fusion genes were introduced into the different loci of chromosomes of S. cerevisiae and expressed under the control of the glyceraldehyde-3-phosphate dehydrogenase promoter. The glucoamylase and alpha-amylase activities were not detected in the culture medium, but in the cell pellet fraction. The transformant strain co-displaying glucoamylase and alpha-amylase could grow faster on starch as the sole carbon source than the transformant strain displaying only glucoamylase.


Subject(s)
Glucan 1,4-alpha-Glucosidase/genetics , Membrane Proteins/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Starch/metabolism , alpha-Amylases/genetics , Agglutinins/genetics , Artificial Gene Fusion , Bacillus/genetics , Cloning, Molecular , Enzyme Repression , Fluorescent Antibody Technique, Indirect , Glucan 1,4-alpha-Glucosidase/biosynthesis , Membrane Proteins/biosynthesis , Recombinant Proteins/biosynthesis , Rhizopus/genetics , Saccharomyces cerevisiae/growth & development , Transfection , alpha-Amylases/biosynthesis
20.
Appl Microbiol Biotechnol ; 48(4): 499-503, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9390459

ABSTRACT

We tried genetically to immobilize cellulase protein on the cell surface of the yeast Saccharomyces cerevisiae in its active form. A cDNA encoding FI-carboxymethylcellulase (CMCase) of the fungus Aspergillus aculeatus, with its secretion signal peptide, was fused with the gene encoding the C-terminal half (320 amino acid residues from the C terminus) of yeast alpha-agglutinin a protein involved in mating and covalently anchored to the cell wall. The plasmid constructed containing this fusion gene was introduced into S. cerevisiae and expressed under the control of the glyceraldehyde-3-phosphate dehydrogenase promoter from S. cerevisiae. The CMCase activity was detected in the cell pellet fraction. The CMCase protein was solubilized from the cell wall fraction by glucanase treatment but not by sodium dodecyl sulphate treatment, indicating the covalent binding of the fusion protein to the cell wall. The appearance of the fused protein on the cell surface was further confirmed by immunofluorescence microscopy and immunoelectron microscopy. These results proved that the CMCase was anchored on the cell wall in its active form.


Subject(s)
Aspergillus/enzymology , Cell Wall/enzymology , Cellulase/metabolism , Enzymes, Immobilized , Fungal Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Cellulase/genetics , Fungal Proteins/genetics , Genes, Synthetic , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Mating Factor , Microscopy, Fluorescence , Microscopy, Immunoelectron , Peptide Fragments/genetics , Peptides/genetics , Peptides/metabolism , Promoter Regions, Genetic , Protein Sorting Signals/genetics , Protein Sorting Signals/metabolism , Saccharomyces cerevisiae/genetics
SELECTION OF CITATIONS
SEARCH DETAIL