Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 153
Filter
1.
Cell Calcium ; 51(5): 351-82, 2012 May.
Article in English | MEDLINE | ID: mdl-22387010

ABSTRACT

The importance of Ca2+-signaling for many subcellular processes is well established in higher eukaryotes, whereas information about protozoa is restricted. Recent genome analyses have stimulated such work also with Alveolates, such as ciliates (Paramecium, Tetrahymena) and their pathogenic close relatives, the Apicomplexa (Plasmodium, Toxoplasma). Here we compare Ca2+ signaling in the two closely related groups. Acidic Ca2+ stores have been characterized in detail in Apicomplexa, but hardly in ciliates. Two-pore channels engaged in Ca2+-release from acidic stores in higher eukaryotes have not been stingently characterized in either group. Both groups are endowed with plasma membrane- and endoplasmic reticulum-type Ca2+-ATPases (PMCA, SERCA), respectively. Only recently was it possible to identify in Paramecium a number of homologs of ryanodine and inositol 1,3,4-trisphosphate receptors (RyR, IP3R) and to localize them to widely different organelles participating in vesicle trafficking. For Apicomplexa, physiological experiments suggest the presence of related channels although their identity remains elusive. In Paramecium, IP3Rs are constitutively active in the contractile vacuole complex; RyR-related channels in alveolar sacs are activated during exocytosis stimulation, whereas in the parasites the homologous structure (inner membrane complex) may no longer function as a Ca2+ store. Scrutinized comparison of the two closely related protozoan phyla may stimulate further work and elucidate adaptation to parasitic life. See also "Conclusions" section.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Inositol Phosphates/physiology , Plasma Membrane Calcium-Transporting ATPases/physiology , Ryanodine Receptor Calcium Release Channel/physiology , Alveolata/physiology , Animals , Biological Evolution , Calcium/chemistry , Genome , Inositol Phosphates/genetics , Paramecium/physiology , Plasma Membrane Calcium-Transporting ATPases/genetics , Plasmodium/physiology , Ryanodine Receptor Calcium Release Channel/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
2.
Eukaryot Cell ; 9(7): 1049-63, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20435698

ABSTRACT

We characterized the calcineurin (CaN) gene family, including the subunits CaNA and CaNB, based upon sequence information obtained from the Paramecium genome project. Paramecium tetraurelia has seven subfamilies of the catalytic CaNA subunit and one subfamily of the regulatory CaNB subunit, with each subfamily having two members of considerable identity on the amino acid level (>or=55% between subfamilies, >or=94% within CaNA subfamilies, and full identity in the CaNB subfamily). Within CaNA subfamily members, the catalytic domain and the CaNB binding region are highly conserved and molecular modeling revealed a three-dimensional structure almost identical to a human ortholog. At 14 members, the size of the CaNA family is unprecedented, and we hypothesized that the different CaNA subfamily members were not strictly redundant and that at least some fulfill different roles in the cell. This was tested by selecting two phylogenetically distinct members of this large family for posttranscriptional silencing by RNA interference. The two targets resulted in differing effects in exocytosis, calcium dynamics, and backward swimming behavior that supported our hypothesis that the large, highly conserved CaNA family members are not strictly redundant and that at least two members have evolved diverse but overlapping functions. In sum, the occurrence of CaN in Paramecium spp., although disputed in the past, has been established on a molecular level. Its role in exocytosis and ciliary beat regulation in a protozoan, as well as in more complex organisms, suggests that these roles for CaN were acquired early in the evolution of this protein family.


Subject(s)
Calcineurin/metabolism , Calcium/metabolism , Catalytic Domain , Multigene Family , Paramecium tetraurelia/enzymology , Protozoan Proteins/metabolism , Calcineurin/genetics , Calcium Signaling/drug effects , Exocytosis/drug effects , Gene Conversion/drug effects , Genes, Protozoan , Introns/genetics , Models, Biological , Movement/drug effects , Mutation/genetics , Paramecium tetraurelia/cytology , Paramecium tetraurelia/drug effects , Paramecium tetraurelia/genetics , Phylogeny , Potassium Chloride/pharmacology , Protein Structure, Tertiary , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , RNA Interference/drug effects , Sequence Homology, Amino Acid , Solutions
3.
J Membr Biol ; 200(1): 15-23, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15386156

ABSTRACT

We analyzed temperature-induced changes of variant surface antigen (vsAg) expression in Paramecium primaurelia, using immuno-techniques and mRNA determinations. Upon a 23 degrees C to 33 degrees C shift, the old vsAg, type 156G, remains on the cell surface for a time, when already mRNA for the new form, 156D, is expressed. A considerable amount of 156D-specific mRNA is formed 45-48 h after the temperature shift, while 156D surface expression reaches maximal levels only after >72 h. A new aspect of these experiments is that, during this transition, the old vsAg is steadily released in high-molecular-weight form into the culture medium, as found by dot blot and Western blot analysis of concentrated culture medium. The new vsAg form is first inserted into the somatic cell membrane, before it spreads also into cilia. In the reverse transition, 33 degrees C to 23 degrees C, the adaptation on the level of transcription and surface expression is considerably faster. While we had previously shown, under steady-state conditions (constant temperature), the occurrence of a degradation pathway by endocytotic and phagocytotic ingestion of vsAg this may proceed in parallel to the steady release of old vsAg from the cell surface into the medium. Altogether these combined processes may facilitate the installation of the new vsAg type.


Subject(s)
Antigens, Protozoan/immunology , Antigens, Surface/immunology , Paramecium/immunology , Temperature , Transcription, Genetic , Animals , Cilia/immunology , Culture Media/chemistry , Microscopy, Immunoelectron , Protozoan Proteins/immunology
4.
Cell Mol Life Sci ; 61(16): 2092-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15316658

ABSTRACT

A new model of caveolin association with lipid body cores has recently been proposed which may be relevant to a number of cellular processes, e.g. lipid body generation. Here we show that PrPc and reggie-1 and reggie-2 also occur in the cores of Nile Red/Bodipy-stained (neutral lipid-containing) vesicular structures and, in immunoblots, in the lipid-enriched fraction after density gradient centrifugation. These lipid-rich vesicles increase in number following cell feeding with oleic acid, differ from early endosome antigen 1- and Lamp-2-positive endosomes/lysosomes, exhibit an opaque content and lack surrounding actin staining. Our results suggest that the content of these vesicles, together with reggie-1 and -2 and PrPc, is expelled.


Subject(s)
Cytoplasmic Vesicles/metabolism , Membrane Proteins/metabolism , PrPC Proteins/metabolism , Actins/metabolism , Cytoplasmic Vesicles/ultrastructure , Fluorescent Dyes , Humans , Immunohistochemistry , Jurkat Cells , Microscopy, Electron , Oxazines , Staining and Labeling
5.
Cell Calcium ; 34(1): 87-96, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12767896

ABSTRACT

This is the first thorough study of refilling of a cortical calcium store in a secretory cell after stimulation in which we combined widely different methodologies. Stimulation of dense-core vesicle ("trichocysts") exocytosis in Paramecium involves a Ca(2+) -influx" superimposed to Ca(2+) -release from cortical stores ("alveolar sacs" (ASs)). In quenched-flow experiments, membrane fusion frequency rose with increasing [Ca(2+)](o) in the medium, from approximately 20-25% at [Ca(2+)](o) < or = 0.25 microM to 100% at [Ca(2+)](o) between 2 and 10 microM, i.e. close to the range of estimated local intracellular [Ca(2+)] during membrane fusion. Next, we analyzed Ca(2+)-specific fluorochrome signals during stimulation under different conditions. Treatment with actin-reactive drugs had no effect on Ca(2+) -signaling. In double trigger experiments, with BAPTA in the second secretagogue application (BAPTA only for stimulation and analysis), the cortical Ca(2+) -signal (due solely to Ca(2+) released from cortical stores) recovered with t(1/2) approximately 65 min. When ASs were analyzed in situ by X-ray microanalysis after different trigger times (+Ca(2+)(o)), t(1/2) for store refilling was similar, approximately 60 min. These values are similar to previously measured 45Ca(2+) -uptake by isolated ASs. In sum we find, (i) exogenous Ca(2+) increases exocytosis/membrane fusion performance with EC(50)=0.7 microM, (ii) Ca(2+) -signaling in this system is not sensitive to actin-reactive drugs, and (iii) refilling of these cortical calcium stores goes on over hours and thus is much slower than expected.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Organelles/metabolism , Paramecium/metabolism , Actins/drug effects , Actins/metabolism , Animals , Calcium/pharmacology , Calcium Signaling/drug effects , Chelating Agents/pharmacology , Cytoplasm/drug effects , Cytoplasm/metabolism , Exocytosis/drug effects , Exocytosis/physiology , Fluorescent Dyes/pharmacology , Intracellular Fluid/drug effects , Intracellular Fluid/metabolism , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Membrane Fusion/drug effects , Membrane Fusion/physiology , Organelles/drug effects , Paramecium/cytology , Paramecium/drug effects , Reaction Time/drug effects , Reaction Time/physiology , Secretory Vesicles/drug effects , Secretory Vesicles/metabolism
6.
J Membr Biol ; 187(1): 1-14, 2002 May 01.
Article in English | MEDLINE | ID: mdl-12029373

ABSTRACT

A non-discharge mutant of Paramecium tetraurelia (nd12-35 degrees C, lacking exocytotic response upon stimulation with the nonpermeable polycationic secretagogue aminoethyldextran, AED), in the pawnA genetic context (d4-500r, lacking ciliary voltage-dependent Ca2+ influx), was shown to lack (45)Ca2+ entry from outside upon AED stimulation. In contrast, cells grown at 25 degrees C behave like the wildtype. To check the functional properties in more detail, fluorochrome-loaded 35 degrees C cells were stimulated, not only with AED (EC(100) = 10(-6) M in wildtype cells), but also with 4-chloro-meta-cresol, (4CmC, 0.5 mM), a permeable activator of ryanodine receptor-type Ca2+ release channels, usually at extracellular [Ca2+] of 50 microM, and eventually with a Ca2+ chelator added. We confirm that pwA-nd12(35 degrees C) cells lack any Ca2+ influx and any exocytosis of trichocysts in response to any stimulus. As we determined by x-ray microanalysis, total calcium content in alveolar sacs (subplasmalemmal stores) known to be mobilized upon exocytosis stimulation in wild-type cells, contain about the same total calcium in 35 degrees C as in 25 degrees C cells, and Ca2+ mobilization from alveoli by AED or 4CmC is also nearly the same. Due to the absence of any AED-induced Ca2+ influx in 35 degrees C cells and normal Ca2+ release from stores found by x-ray microanalysis one can exclude a "CICR"-type mechanism (Ca2+-induced Ca2+ release) and imply that normally a store-operated Ca2+ ("SOC") influx would occur (as in 25 degrees C cells). Furthermore, 35 degrees C cells display a significantly lower basal intracellular [Ca2+], so that any increase upon stimulation may be less expressed or even remain undetected. Under these conditions, any mobilization of Ca2+ from stores cannot compensate for the lack of Ca2+ influx, particularly since normally both components have to cooperate to achieve full exocytotic response. Also striking is our finding that 35 degrees C cells are unable to perform membrane fusion, as analyzed with the Ca2+ ionophore, A23187. These findings were corroborated by cryofixation and freeze-fracture analysis of trichocyst docking sites after AED or 4CmC stimulation, which also revealed no membrane fusion. In sum, in nd12 cells increased culture temperature entails multiple defects, notably insensitivity to any Ca2+ signal, which, moreover, cannot develop properly due to a lower basal [Ca2+] level and the lack of Ca2+ influx, despite normal store activation.


Subject(s)
Calcium/metabolism , Cresols/metabolism , Dextrans/metabolism , Exocytosis/physiology , Paramecium tetraurelia/genetics , Paramecium tetraurelia/metabolism , Animals , Caffeine/metabolism , Caffeine/pharmacology , Calcium Channels/drug effects , Cell Line , Cresols/pharmacology , Dextrans/pharmacology , Exocytosis/drug effects , Fluorescent Dyes/metabolism , Mutation , Paramecium tetraurelia/drug effects , Paramecium tetraurelia/ultrastructure , Sensitivity and Specificity
7.
Mol Biol Cell ; 12(10): 3031-45, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11598189

ABSTRACT

Using confocal laser scanning and double immunogold electron microscopy, we demonstrate that reggie-1 and -2 are colocalized in < or =0.1-microm plasma membrane microdomains of neurons and astrocytes. In astrocytes, reggie-1 and -2 do not occur in caveolae but clearly outside these structures. Microscopy and coimmunoprecipitation show that reggie-1 and -2 are associated with fyn kinase and with the glycosylphosphatidyl inositol-anchored proteins Thy-1 and F3 that, when activated by antibody cross-linking, selectively copatch with reggie. Jurkat cells, after cross-linking of Thy-1 or GM1 (with the use of cholera toxin), exhibit substantial colocalization of reggie-1 and -2 with Thy-1, GM1, the T-cell receptor complex and fyn. This, and the accumulation of reggie proteins in detergent-resistant membrane fractions containing F3, Thy-1, and fyn imparts to reggie-1 and -2 properties of raft-associated proteins. It also suggests that reggie-1 and -2 participate in the formation of signal transduction centers. In addition, we find reggie-1 and -2 in endolysosomes. In Jurkat cells, reggie-1 and -2 together with fyn and Thy-1 increase in endolysosomes concurrent with a decrease at the plasma membrane. Thus, reggie-1 and -2 define raft-related microdomain signaling centers in neurons and T cells, and the protein complex involved in signaling becomes subject to degradation.


Subject(s)
Fish Proteins , Glycosylphosphatidylinositols/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins , Nerve Tissue Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Thy-1 Antigens/metabolism , Animals , Astrocytes/metabolism , Brain/metabolism , Caveolae/metabolism , Caveolae/ultrastructure , Caveolins/metabolism , Cell Adhesion Molecules/metabolism , Cell Membrane/metabolism , G(M1) Ganglioside/metabolism , Humans , Immunohistochemistry , Jurkat Cells/metabolism , Lysosomes/metabolism , Membrane Proteins/ultrastructure , Microscopy, Confocal , Nerve Tissue Proteins/ultrastructure , PC12 Cells/metabolism , Protein Structure, Tertiary/physiology , Rats , Signal Transduction/physiology , src-Family Kinases
8.
Trends Genet ; 17(6): 306-8, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11377780

ABSTRACT

A consortium of laboratories undertook a pilot sequencing project to gain insight into the genome of Paramecium. Plasmid-end sequencing of DNA fragments from the somatic nucleus together with similarity searches identified 722 potential protein-coding genes. High gene density and uniform small intron size make random sequencing of somatic chromosomes a cost-effective strategy for gene discovery in this organism.


Subject(s)
Genome, Protozoan , Paramecium/genetics , Animals , Humans , Paramecium/classification , Phylogeny , Pilot Projects , Protozoan Proteins/genetics
9.
Int Rev Cytol ; 201: 115-208, 2001.
Article in English | MEDLINE | ID: mdl-11057832

ABSTRACT

In ciliates, a variety of processes are regulated by Ca2+, e.g., exocytosis, endocytosis, ciliary beat, cell contraction, and nuclear migration. Differential microdomain regulation may occur by activation of specific channels in different cell regions (e.g., voltage-dependent Ca2+ channels in cilia), by local, nonpropagated activation of subplasmalemmal Ca stores (alveolar sacs), by different sensitivity thresholds, and eventually by interplay with additional second messengers (cilia). During stimulus-secretion coupling, Ca2+ as the only known second messenger operates at approximately 5 microM, whereby mobilization from alveolar sacs is superimposed by "store-operated Ca2+ influx" (SOC), to drive exocytotic and endocytotic membrane fusion. (Content discharge requires binding of extracellular Ca2+ to some secretory proteins.) Ca2+ homeostasis is reestablished by binding to cytosolic Ca2+-binding proteins (e.g., calmodulin), by sequestration into mitochondria (perhaps by Ca2+ uniporter) and into endoplasmic reticulum and alveolar sacs (with a SERCA-type pump), and by extrusion via a plasmalemmal Ca2+ pump and a Na+/Ca2+ exchanger. Comparison of free vs total concentration, [Ca2+] vs [Ca], during activation, using time-resolved fluorochrome analysis and X-ray microanalysis, respectively, reveals that altogether activation requires a calcium flux that is orders of magnitude larger than that expected from the [Ca2+] actually required for local activation.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Ciliophora/cytology , Ciliophora/metabolism , Animals , Cell Division/physiology , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Movement/physiology , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cilia/metabolism , Contractile Proteins/metabolism , Endocytosis/physiology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Eukaryotic Cells/cytology , Eukaryotic Cells/metabolism , Exocytosis/physiology , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Ion Channels/metabolism , Ion Channels/ultrastructure , Secretory Vesicles/metabolism , Secretory Vesicles/ultrastructure
10.
J Cell Biol ; 151(3): 519-28, 2000 Oct 30.
Article in English | MEDLINE | ID: mdl-11062254

ABSTRACT

Many intracellular compartments of eukaryotic cells do not adopt a spherical shape, which would be expected in the absence of mechanisms organizing their structure. However, little is known about the principles determining the shape of organelles. We have observed very defined structural changes of vacuoles, the lysosome equivalents of yeast. The vacuolar membrane can form a large tubular invagination from which vesicles bud off into the lumen of the organelle. Formation of the tube is regulated via the Apg/Aut pathway. Its lumen is continuous with the cytosol, making this inverse budding reaction equivalent to microautophagocytosis. The tube is highly dynamic, often branched, and defined by a sharp kink of the vacuolar membrane at the site of invagination. The tube is formed by vacuoles in an autonomous fashion. It persists after vacuole isolation and, therefore, is independent of surrounding cytoskeleton. There is a striking lateral heterogeneity along the tube, with a high density of transmembrane particles at the base and a smooth zone devoid of transmembrane particles at the tip where budding occurs. We postulate a lateral sorting mechanism along the tube that mediates a depletion of large transmembrane proteins at the tip and results in the inverse budding of lipid-rich vesicles into the lumen of the organelle.


Subject(s)
Endocytosis , Intracellular Membranes/metabolism , Phagocytosis , Saccharomyces cerevisiae/cytology , Vacuoles/metabolism , Vacuoles/ultrastructure , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Cytosol/metabolism , Cytosol/ultrastructure , Freeze Fracturing , Intracellular Membranes/ultrastructure , Lysosomes/chemistry , Lysosomes/metabolism , Lysosomes/ultrastructure , Membrane Fusion , Microscopy, Electron , Microscopy, Fluorescence , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/ultrastructure , Vacuoles/chemistry
11.
Eur J Cell Biol ; 79(9): 642-52, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11043405

ABSTRACT

Though only actual local free Ca2+ concentrations, [Ca2+], rather than total Ca concentrations, [Ca], govern cellular responses, analysis of total calcium fluxes would be important to fully understand the very complex Ca2+ dynamics during cell stimulation. Using Paramecium cells we analyzed Ca2+ mobilization from cortical stores during synchronous (< or = 80 ms) exocytosis stimulation, by quenched-flow/cryofixation, freeze-substitution (modified for Ca retention) and X-ray microanalysis which registers total calcium concentrations, [Ca]. When the extracellular free calcium concentration, [Ca2+]e, is adjusted to approximately 30 nM, i.e. slightly below the normal free intracellular calcium concentration, [Ca2+]i = 65 nM, exocytosis stimulation causes release of 52% of calcium from stores within 80 ms. At higher extracellular calcium concentration, [Ca2+]e = 500 microM, Ca2+ release is counterbalanced by influx into stores within the first 80 ms, followed by decline of total calcium, [Ca], in stores to 21% of basal values within 1 s. This includes the time required for endocytosis coupling (350 ms), another Ca2+-dependent process. To confirm that Ca2+ mobilization from stores is superimposed by rapid Ca2+ influx and/or uptake into stores, we substituted Sr2+ for Ca2+ in the medium for 500 ms, followed by 80 ms stimulation. This reveals reduced Ca signals, but strong Sr signals in stores. During stimulation, Ca2+ is spilled over preformed exocytosis sites, particularly with increasing extracellular free calcium, [Ca2+]e. Cortically enriched mitochondria rapidly gain Ca signals during stimulation. Balance calculations indicate that total Ca2+ flux largely exceeds values of intracellular free calcium concentrations locally required for exocytosis (as determined previously). Our approach and some of our findings appear relevant also for some other secretory systems.


Subject(s)
Calcium/metabolism , Electron Probe Microanalysis/methods , Exocytosis/physiology , Microscopy, Electron, Scanning Transmission/methods , Paramecium tetraurelia/metabolism , Animals , Calcium Signaling/physiology , Chelating Agents/pharmacology , Cilia/metabolism , Cilia/ultrastructure , Cytosol/metabolism , Cytosol/ultrastructure , Egtazic Acid/pharmacology , Exocytosis/drug effects , Freeze Substitution , Mitochondria/metabolism , Mitochondria/ultrastructure , Strontium/pharmacokinetics
12.
Mol Microbiol ; 37(4): 773-87, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10972800

ABSTRACT

We have followed the time-dependent transfection of Paramecium cells with a vector containing the gene of green fluorescent protein (GFP) attached to the C-terminus of the PtSERCA1 gene. The outlines of alveolar sacs (ASs) are labelled, as is the endoplasmic reticulum (ER) throughout the cell. When GFP fluorescence is compared with previous anti-PtSERCA1 antibody labelling, the much wider distribution of GFP (ER+ASs) indicates that only a small amount of SERCA molecules is normally retained in the ER. A second isoform, PtSERCA2, also occurs and its C-terminal GFP-tagging results in the same distribution pattern. However, when GFP is inserted in the major cytoplasmic loop, PtSERCA1 and two fusion proteins are mostly retained in the ER, probably because of the presence of the overt C-terminal KKXX ER-retention signal and/or masking of a signal for transfer into ASs. On the overall cell surface, new SERCA molecules seem to be permanently delivered from the ER to ASs by vesicle transport, whereas in the fission zone of dividing cells ASs may form anew. In cells overexpressing PtSERCA1 (with C-terminal GFP) in ASs, [Ca2+]i regulation during exocytosis is not significantly different from controls, probably because their Ca2+ pump has to mediate only slow reuptake.


Subject(s)
Calcium-Transporting ATPases/metabolism , Calcium/metabolism , Endoplasmic Reticulum/enzymology , Isoenzymes/metabolism , Paramecium/genetics , Sarcoplasmic Reticulum/enzymology , Subcellular Fractions/enzymology , Amino Acid Sequence , Animals , Base Sequence , Calcium-Transporting ATPases/genetics , DNA Primers , Endoplasmic Reticulum/ultrastructure , Green Fluorescent Proteins , Isoenzymes/genetics , Luminescent Proteins/genetics , Microscopy, Confocal , Microscopy, Electron , Sarcoplasmic Reticulum/ultrastructure
13.
J Histochem Cytochem ; 48(9): 1269-81, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10950883

ABSTRACT

For immunogold EM labeling analysis, we fixed Paramecium cells in 4% formaldehyde and 0.125% glutaraldehyde, followed by low-temperature embedding in unicryl and UV polymerization. We first quantified some obvious but thus far neglected side effects of section staining on immunogold labeling, using mono- or polyclonal antibodies (Abs) against defined secretory and cell surface components, followed by F(ab)(2)- or protein A-gold conjugates. Use of alkaline lead staining resulted in considerable rearrangement and loss of label unless sections were postfixed by glutaraldehyde after gold labeling. This artifact is specific for section staining with lead. It can be avoided by staining sections with aqueous uranyl acetate only to achieve high-resolution immunogold localization of a protein phosphatase on unicryl sections. In general, phosphatases are assumed to be closely, although loosely, associated with their targets. Because the occurrence of protein phosphatase 2B (calcineurin) in Paramecium has been previously established by biochemical and immunological work, as well as by molecular biology, we have used Abs against mammalian CaN or its subunits, CaN-A and CaN-B, for antigen mapping in these cells by quantitative immunogold labeling analysis. Using ABs against whole CaN, four structures are selectively labeled (with slightly decreasing intensity), i.e., infraciliary lattice (centrin-containing contractile cortical filament network), parasomal sacs (coated pits), and outlines of alveolar sacs (subplasmalemmal calcium stores, tightly attached to the cell membrane), as well as rims of chromatin-containing nuclear domains. In other subcellular regions, gold granules reached densities three to four times above background outside the cell but there was no selective enrichment, e.g., in cilia, ciliary basal bodies, cytosol, mitochondria, trichocysts (dense-core secretory organelles), and non-chromatin nuclear domains. Their labeling density was 4- to 8.5-fold (average 6.5-fold) less than that on selectively labeled structures. Labeling tendency was about the same with Abs against either subunit. Our findings may facilitate the examination of molecular targets contained in the selectively labeled structures. (J Histochem Cytochem 48:1269-1281, 2000)


Subject(s)
Calcineurin/metabolism , Paramecium/enzymology , Animals , Antibodies , Artifacts , Blotting, Western , Calcineurin/immunology , Immunohistochemistry/methods , Lead , Paramecium/cytology , Paramecium/ultrastructure , Tissue Fixation
14.
J Membr Biol ; 176(3): 237-48, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10931975

ABSTRACT

The lipophilic fluorescent dye, FM1-43, as now frequently used to stain cell membranes and to monitor exo-endocytosis and membrane recycling, induces a cortical [Ca(2+)](i) transient and exocytosis of dense core vesicles ("trichocysts") in Paramecium cells, when applied at usual concentrations (

Subject(s)
Exocytosis , Membrane Fusion/physiology , Paramecium tetraurelia/physiology , Animals , Calcium/metabolism , Calcium/pharmacology , Calmodulin/antagonists & inhibitors , Calmodulin/physiology , Fluorescent Dyes/metabolism , Imidazoles/pharmacology , Ion Transport , Magnesium/pharmacology , Membrane Fusion/drug effects , Organelles/physiology , Organelles/ultrastructure , Paramecium tetraurelia/drug effects , Paramecium tetraurelia/ultrastructure , Pyridinium Compounds/metabolism , Quaternary Ammonium Compounds/metabolism
15.
J Membr Biol ; 174(2): 141-56, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10742458

ABSTRACT

The polyamine secretagogue, aminoethyldextran (AED), causes a cortical [Ca(2+)] transient in Paramecium cells, as analyzed by fluorochrome imaging. Our most essential findings are: (i) Cortical Ca(2+) signals also occur when AED is applied in presence of the fast Ca(2+) chelator, BAPTA. (ii) Extracellular La(3+) application causes within seconds a rapid, reversible fluorescence signal whose reversibility can be attributed to a physiological [Ca(2+)](i) transient (while injected La(3+) causes a sustained fluorescence signal). (iii) Simply increasing [Ca(2+)](o) causes a similar rapid, short-lived [Ca(2+)](i) transient. All these phenomena, (i-iii), are compatible with activation of an extracellular "Ca(2+)/(polyvalent cation)-sensing receptor" known from some higher eukaryotic systems, where this sensor (responding to Ca(2+), La(3+) and some multiply charged cations) is linked to cortical calcium stores which, thus, are activated. In Paramecium, such subplasmalemmal stores ("alveolar sacs") are physically linked to the cell membrane and they can also be activated by the Ca(2+) releasing agent, 4-chloro-m-cresol, just like in Sarcoplasmic Reticulum. Since this drug causes a cortical Ca(2+) signal also in absence of Ca(2+)(o) we largely exclude a "Ca(2+)-induced Ca(2+) release" (CICR) mechanism. Our finding of increased cortical Ca(2+) signals after store depletion and re-addition of extracellular Ca(2+) can be explained by a "store-operated Ca(2+) influx" (SOC), i.e., a Ca(2+) influx superimposing store activation. AED stimulation in presence of Mn(2+)(o) causes fluorescence quenching in Fura-2 loaded cells, indicating involvement of unspecific cation channels. Such channels, known to occur in Paramecium, share some general characteristics of SOC-type Ca(2+) influx channels. In conclusion, we assume the following sequence of events during AED stimulated exocytosis: (i) activation of an extracellular Ca(2+)/polyamine-sensing receptor, (ii) release of Ca(2+) from subplasmalemmal stores, (iii) and Ca(2+) influx via unspecific cation channels. All three steps are required to produce a steep cortical [Ca(2+)] signal increase to a level required for full exocytosis activation. In addition, we show formation of [Ca(2+)] microdomains (

Subject(s)
Calcium/metabolism , Dextrans/metabolism , Ion Channels/metabolism , Paramecium/metabolism , Receptors, Cell Surface/metabolism , Animals , Calcium Channels/metabolism , Dextrans/pharmacology , Exocytosis/physiology , Extracellular Space , Lanthanum/metabolism , Paramecium/drug effects , Polyamines/pharmacology , Receptors, Calcium-Sensing
16.
Eur J Cell Biol ; 79(2): 144-9, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10727022

ABSTRACT

In this paper we describe the expression of green fluorescent protein (GFP) as a reporter in vivo to monitor transformation in Paramecium cells. This is not trivial because of the limited number of strong promoters available for heterologous expression and the very high AT content of the genomic DNA, the consequence of which is a very aberrant codon usage. Taking into account differences in codon usage we selected and modified the original GFP open reading frame (ORF) from Aequorea victoria and placed the altered ORF into the Paramecium expression vector pPXV. Injection of the linearized plasmid into the macronucleus resulted in a cytoplasmic fluorescence signal in the clonal descendants, which was proportional to the number of copies injected. Southern hybridization indicated the establishment and replication of the plasmid during vegetative growth. Expression was also monitored by Northern and Western analysis. The results indicate that the modified GFP can be used in Paramecium as a reporter for transformation as an alternative to selection with antibiotics and that it may also be used to construct and localize fusion proteins.


Subject(s)
Genes, Reporter , Luminescent Proteins/genetics , Paramecium tetraurelia/genetics , Animals , Gene Expression , Gene Transfer Techniques , Green Fluorescent Proteins , Luminescent Proteins/biosynthesis , Paramecium tetraurelia/metabolism , Promoter Regions, Genetic
17.
J Struct Biol ; 128(2): 187-99, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10600572

ABSTRACT

We analyzed preparative and analytical aspects of the dynamic localization of Ca(2+) during cell stimulation, using a combination of quenched flow and energy-dispersive X-ray microanalysis (EDX). Calcium (or Sr, as a substitute) was retained as fluorides during freeze-substitution, followed by epoxide embedding. The quenched-flow used allowed analyses, during stimulation, in the subsecond time range. Sections of 500 nm were analyzed and no artificial Ca or Sr leakage was recognizable. We calculated a primary beam spread from 63 to 72 nm that roughly indicated the resolution of EDX/structure correlation. These values are quite compatible with the size of potential structures of interest, e.g., Ca stores (approximately 100-nm thickness) or cilia (approximately 250-nm diameter). We used widely different standards to calibrate the ratio of CaK(alpha) net counts in relation to actual ¿Ca. Calibration curves showed a linear relationship and a detection limit of ¿Ca = 2 mM, while ¿Ca in cytosol was 3 mM and in stores was 43 mM, both in nonactivated cells. Eventually Sr(2+) can rapidly be substituted for Ca(2+) in the medium before and during stimulation, thus allowing one to determine Me(2+) fluxes. With our "model" cell, Paramecium, we showed that, upon stimulation (causing rapid Ca(2+) mobilization from subplasmalemmal stores), Ca was immediately exchanged for Sr in stores.


Subject(s)
Calcium/analysis , Paramecium tetraurelia/metabolism , Animals , Cilia/ultrastructure , Cryopreservation , Dextrans/pharmacology , Electron Probe Microanalysis , Exocytosis , Microscopy, Electron, Scanning Transmission , Paramecium tetraurelia/ultrastructure , Polystyrenes , Polyvinyls , Sensitivity and Specificity
18.
J Membr Biol ; 172(1): 77-88, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10552016

ABSTRACT

We combined widely different biochemical methods to analyze proteins of the cell surface of P. tetraurelia since so far one can isolate only a subfraction of cell membrane vesicles enriched in the GPI-anchored surface antigens ("immoblization" or "i-AGs"). We also found that i-AGs may undergo partial degradation by endogenous proteases. Genuine intrinsic membrane proteins were recognized particularly with lipophilic 5-[(125)I]-iodonaphthalene-1-azide (INA) labeling which reportedly "sees" integral proteins and cytoplasmic cell membrane-associated proteins. With INA (+DTT), bands of

Subject(s)
Paramecium/chemistry , Protozoan Proteins/isolation & purification , Animals , Antigens, Protozoan/chemistry , Antigens, Protozoan/isolation & purification , Antigens, Surface/chemistry , Antigens, Surface/isolation & purification , Azides , Cell Membrane/chemistry , Cholic Acids , Cross-Linking Reagents , Iodine Radioisotopes , Membrane Proteins/chemistry , Membrane Proteins/immunology , Membrane Proteins/isolation & purification , Molecular Weight , Paramecium/immunology , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Solubility
19.
EMBO J ; 18(15): 4222-32, 1999 Aug 02.
Article in English | MEDLINE | ID: mdl-10428960

ABSTRACT

We cloned a guanylyl cyclase of 280 kDa from the ciliate Paramecium which has an N-terminus similar to that of a P-type ATPase and a C-terminus with a topology identical to mammalian adenylyl cyclases. Respective signature sequence motifs are conserved in both domains. The cytosolic catalytic C1a and C2a segments of the cyclase are inverted. Genes coding for topologically identical proteins with substantial sequence similarities have been cloned from Tetrahymena and were detected in sequences from Plasmodium deposited by the Malaria Genome Project. After 99 point mutations to convert the Paramecium TAA/TAG-Gln triplets to CAA/CAG, together with partial gene synthesis, the gene from Paramecium was heterologously expressed. In Sf9 cells, the holoenzyme is proteolytically processed into the two domains. Immunocytochemistry demonstrates expression of the protein in Paramecium and localizes it to cell surface membranes. The data provide a novel structural link between class III adenylyl and guanylyl cyclases and imply that the protozoan guanylyl cyclases evolved from an ancestral adenylyl cyclase independently of the mammalian guanylyl cyclase isoforms. Further, signal transmission in Ciliophora (Paramecium, Tetrahymena) and in the most important endoparasitic phylum Apicomplexa (Plasmodium) is, quite unexpectedly, closely related.


Subject(s)
Guanylate Cyclase/chemistry , Isoenzymes/chemistry , Paramecium/enzymology , Plasmodium/enzymology , Tetrahymena/enzymology , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA Primers , Guanylate Cyclase/genetics , Humans , Isoenzymes/genetics , Mammals , Microscopy, Immunoelectron , Molecular Sequence Data , Mutagenesis, Site-Directed , Paramecium/genetics , Paramecium/ultrastructure , Plasmodium/genetics , Protein Conformation , Sequence Homology, Amino Acid , Tetrahymena/genetics
20.
Biochemistry ; 38(24): 7780-90, 1999 Jun 15.
Article in English | MEDLINE | ID: mdl-10387018

ABSTRACT

PP63 (parafusin) is a 63 kDa phosphoprotein, which exists in at least two different isoforms. It is very rapidly (80 ms) dephosphorylated during triggered trichocyst exocytosis. This occurs selectively in exocytosis-competent Paramecium tetraurelia strains. At least two protein kinases isolated from Paramecium, casein kinase type II kinase and cGMP-dependent kinase, are able to phosphorylate the two recombinant PP63/parafusin isoforms, both with phosphoglucomutase activity, in vitro. By performing mass spectrometric peptide mapping, we have investigated in vitro phosphorylation of recombinant PP63/parafusin by these kinases in comparison to in vivo phosphorylation of native PP63/parafusin isolated from Paramecium homogenates. Low picomolar quantities of proteolytic digests of recombinant and native PP63/parafusin, prior to and following alkaline phosphatase treatment, were directly analyzed by MALDI mass spectrometry. In native PP63-1/parafusin-1, six of 64 serine and threonine residues (S-196, T-205, T-280, T-371, T-373, and T-469) were found definitely, 27 were found possibly phosphorylated, 28 were identified as nonphosphorylated, and three were not covered by mapping. Three of the six certainly phosphorylated amino acids represent consensus phosphorylation sites for casein kinase II or cGMP-dependent protein kinase. In vitro phosphorylation studies of recombinant PP63/parafusin confirm that some of the sites found were used in vivo; however, also significant differences with respect to in vivo phosphorylation of native PP63/parafusin were observed. The two Paramecium protein kinases that were used do not preferably phosphorylate expected consensus sites in vitro. Homology structure modeling of PP63/parafusin with rabbit phosphoglucomutase revealed that the majority of residues found phosphorylated is located on the surface of the molecule.


Subject(s)
Exocytosis , Paramecium tetraurelia/metabolism , Peptide Mapping , Phosphoglucomutase , Phosphoproteins/metabolism , Protozoan Proteins/metabolism , Amino Acid Sequence , Animals , Casein Kinase II , Cells, Cultured , Cyclic GMP-Dependent Protein Kinases/metabolism , Histidine/genetics , Models, Molecular , Molecular Sequence Data , Paramecium tetraurelia/cytology , Peptide Mapping/methods , Phosphoproteins/chemistry , Phosphoproteins/genetics , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
SELECTION OF CITATIONS
SEARCH DETAIL
...