scholarly journals Radixin is a novel member of the band 4.1 family.

1991 ◽  
Vol 115 (4) ◽  
pp. 1039-1048 ◽  
Author(s):  
N Funayama ◽  
A Nagafuchi ◽  
N Sato ◽  
S Tsukita ◽  
S Tsukita

Radixin is an actin barbed-end capping protein which is highly concentrated in the undercoat of the cell-to-cell adherens junction and the cleavage furrow in the interphase and mitotic phase, respectively (Tsukita, Sa., Y. Hieda, and Sh. Tsukita. 1989 a.J. Cell Biol. 108:2369-2382; Sato, N., S. Yonemura, T. Obinata, Sa. Tsukita, and Sh. Tsukita. 1991. J. Cell Biol. 113:321-330). To further understand the structure and functions of the radixin molecule, we isolated and sequenced the cDNA clones encoding mouse radixin. Direct peptide sequencing of radixin and immunological analysis with antiserum to a fusion protein were performed to confirm that the protein encoded by these clones is identical to radixin. The composite cDNA is 4,241 nucleotides long and codes for a 583-amino acid polypeptide with a calculated molecular mass of 68.5 kD. Sequence analysis has demonstrated that mouse radixin shares 75.3% identity with human ezrin, which was reported to be a member of the band 4.1 family. We then isolated the cDNA encoding mouse ezrin. Sequence analysis and Northern blot analysis revealed that radixin and ezrin are similar but distinct (74.9% identity), leading us to conclude that radixin is a novel member of the band 4.1 family. In erythrocytes the band 4.1 protein acts as a key protein in the association of short actin filaments with a plasma membrane protein (glycophorin), together with spectrin. Therefore, the sequence similarity between radixin and band 4.1 protein described in this study favors the idea that radixin plays a crucial role in the association of the barbed ends of actin filaments with the plasma membrane in the cell-to-cell adherens junction and the cleavage furrow.

1993 ◽  
Vol 105 (2) ◽  
pp. 589-600 ◽  
Author(s):  
T. Maekawa ◽  
R. Kuriyama

Using a human autoantibody, SP-H, we identified a 200–230 kDa mitotic MAP in a variety of mammalian cell lines which shows affinity for the minus end of microtubules and also becomes associated with the spindle pole during mitosis. To examine the detailed structure and functional organization of the protein, the gene coding for the end-specific MAP was isolated and characterized by screening a human placenta lambda gt11 expression library using SP-H as a probe. Overlapping cDNA clones, which covered the entire length of the coding region of the SP-H antigen, were obtained. Polyclonal antibodies raised against fusion proteins generated from non-overlapping cDNA fragments stained the HeLa SP-H antigen in interphase and mitotic cells, and recognized a single 215 kDa band on immunoblots, as did the original SP-H antibody. Analysis of the nucleotide sequence revealed a 7,091 nucleotide sequence with an open reading frame of 6,345 nucleotides encoding a 2,115 amino acid polypeptide with a calculated molecular mass of 238,376 Da. The predicted amino acid sequence showed the protein to be composed of an alpha-helical domain, flanked by globular domains located at the amino and carboxy termini. The sequence contained five repeats of the hypothetical leucine zipper motif: one is in the N-terminal globular domain, and four are in the central alpha-helical stalk. Comparison with other sequences in the database shows that the SP-H antigen is identical to the NuMA protein reported by Yang et al. (1992) J. Cell Biol. 116, 1303–1317, but there are differences between the SP-H antigen and NuMA sequence reported by Compton et al. (1992) J. Cell Biol. 116, 1395–1408. cDNA inserts of the truncated SP-H antigen were expressed in both insect Sf9 cells and in cultured mammalian cells. The recombinant protein corresponding to the C-terminal half of the protein was restricted to the nucleus, whereas the N-terminal half of the protein was localized in the cytoplasm, suggesting the presence of a nuclear translocation signal(s) in the C-terminal domain. The C-terminal polypeptide expressed in mitotic COS cells was shown to specifically localize at the spindle pole. Microtubule-binding assays using in vitro transcribed/translated polypeptide products from different domains of the SP-H antigen further suggested that the SP-H antigen interacts with microtubules through the globular domain at the C-terminus.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1472-1472
Author(s):  
Shugo Kowata ◽  
Tatsuo Oyake ◽  
Shigeki Ito ◽  
Kazunori Murai ◽  
Yoji Ishida

Abstract Abstract 1472 Poster Board I-495 Platelets are generated from the proplatelets of mature megakaryocyte (MK) in bone marrow (BM). Proplatelet formation (PPF) requires profound changes in the cytoskeletal elements including microtubules and actin fibers. The bending and branching of proplatelet shafts are mediated by actin dynamics (Italiano JE Jr, J Cell Biol, 1999) in the process of PPF. Recent reports showed that Rho and its effecter ROCK, inhibit PPF (Chang Y. et al, Blood, 2007) and that the Wiskott-Aldrich syndrome protein (N-WASP) and its effector Arp2/3 complex, the actin nucleating factor, are essential in PPF (Schulze H. et al Blood, 2006). The contribution of another actin nucleating factor, mammalian diaphanous-related forming (mDia1), which is the other downstream effector of Rho, however, has not been reported in the process of PPF. In this study, we investigated the role of these factors and free barbed ends of actin filaments during PPF using the immunofluorescence method and inhibitory assay with specific inhibitors. Materials and methods. Inhibitory assay: Primary mature MKs, isolated by albumin density gradient method, were cultured in IMDM supplemented with 1% Insulin-Transferin-Selenium (ITS) with the specific signal inhibitor of each signal pathway, including Rho, Rho kinase (ROCK), Rac1 or N-WASP. After incubation at 37°C in 5%CO2 and 20%O2 for 16 hrs, MKs were fixed with 4% paraformaldehyde and then counted the ratio of MKs with PPF. Immunofluorescence method: Isolated MKs were cultured in the same condition, described above, fixed with 4% paraformaldehyde and incubated with the following primary antibodies: anti-mDia1 antibody, anti-Arp3 antibody for 1 hr. After washing, MKs were stained with fluolescence conjugated-antibody against the primary antibody. The free barbed end assay: To allow the visualization of actin nucleation sites, it was performed as described previously (Symons and Mitchison, J Cell Biol, 1991). Fluorescence in MKs was observed under a Zeiss LSM meta confocal microscopy. Results and Discussions. Inhibitory assay: The ratio of MKs with PPF increased significantly at low concentration of cell permeable C3 transferase, the specific inhibitor of Rho, (5 ug/ml)(38.4±7.2%, Control 20.3±8.8%, p<0.05) but decreased significantly at high concentration (data not shown). These data indicated that only ROCK might be inactivated at 5 ug/ml but both ROCK and mDia1 might be inactivated at high concentration. The ratio of MKs with PPF increased in the presence of Y27632, a specific inhibitor of ROCK (10 uM: 51.1±10.2%, Control 20.3±8.8%, p<0.05). However, inhibitor of Rac1 or N-WASP signaling decreased the ratio of MKs with PPF compared to the controls (NSC23766: 50 uM: 8.0±5.2%. Wiskostatin: 50uM: 4±3.4% vs control 18.3±6.8%, p<0.05). These data indicated that Arp2/3 was an important role in PPF, because Rac-1 and N-WASP were located at the upperstream of Arp2/3. Immunofluorescence method & free barbed end assay: Isolated MKs without PPF were round and smooth on their surface, and had a thin F-actin layer under the surface area. Free barbed end (FBE) signals were clearly observed along the plasma membrane and co-localized with mDia1 signals (Figure 1), not with Arp3 signal (data not shown). Thesedata indicated the presence of dynamic equilibrium of actin filaments, regulated by Rho/mDia1 signal, around the plasma membrane of MKs. Considering the previous reports in which Rho/Rock/myosin light chain/myosin IIA pathway was reported to restrain PPF and inhibit releasing premature platelets until the appropriate time, myosin IIA might utilize the actin filaments which was produced via Rho/mDia1 signal. In proplatelets, FBE were accumulated in the roots of proplatelet shaft, bifurcation sites, thin filopodias, and beads (Figure 2). These data indicated that actin filaments, produced at FBE, might have important roles in leading of microtubules at the roots and bifrucation sites and beads formation. mDia1 and Arp3 signals were localized at these sites. The differences of localization between mDia1 and Arp3 signals were that only mDia1 signals were accumulated at thin filopodias around the shaft and bead (data not shown). In conclusion, actin dynamics, which is controlled by the actin nucleating factor Arp3 and mDia1 play important roles in PPF of MKs. Disclosures: No relevant conflicts of interest to declare.


1984 ◽  
Vol 99 (1) ◽  
pp. 95s-103s ◽  
Author(s):  
P Mangeat ◽  
K Burridge

In this review we discuss some of the proteins for which a role in linking actin to the fibroblast plasma membrane has been suggested. We focus on the family of proteins related to erythrocyte spectrin, proteins that have generally been viewed as having an organization and a function in actin-membrane attachment similar to those of erythrocyte spectrin. Experiments in which we precipitated the nonerythrocyte spectrin within living fibroblasts have led us to question this supposed similarity of organization and function of the nonerythrocyte and erythrocyte spectrins. Intracellular precipitation of fibroblast spectrin does not affect the integrity of the major actin-containing structures, the stress fiber microfilament bundles. Unexpectedly, however, we found that the precipitation of spectrin results in a condensation and altered distribution of the vimentin class of intermediate filaments in most cells examined. Although fibroblast spectrin may have a role in the attachment of some of the cortical, submembranous actin, it is surprising how little the intracellular immunoprecipitation of the spectrin affects the cells. Several proteins have been found concentrated at the ends of stress fibers, where the actin filaments terminate at focal contacts. Two of these proteins, alpha-actinin and fimbrin, have properties that suggest that they are not involved in the attachment of the ends of the bundles to the membrane but are more probably involved in the organization and cross-linking of the filaments within the bundles. On the other hand, vinculin and talin are two proteins that interact with each other and may form part of a chain of attachments between the ends of the microfilament bundles and the focal contact membrane. Their role in this attachment, however, has not been established and further work is needed to examine their interaction with actin and to identify any other components with which they may interact, particularly in the plasma membrane.


1976 ◽  
Vol 69 (1) ◽  
pp. 51-72 ◽  
Author(s):  
LG Tilney

At an early stage in spermiogenesis the acrosomal vacuole and other organelles including ribosomes are located at the basal end of the cell. From here actin must be transported to its future location at the anterior end of the cell. At no stage in the accumulation of actin in the periacrosomal region is the actin sequestered in a membrane-bounded compartment such as a vacuole or vesicle. Since filaments are not present in the periacrosomal region during the accumulation of the actin even though the fixation of these cells is sufficiently good to distinguish actin filaments in thin section, the actin must accumulate in the nonfilamentous state. The membranes in the periacrosomal region, specifically a portion of the nuclear envelope and the basal half of the acrosomal vacuole membrane, become specialized morphologically in advance of the accumulation of actin in this region. My working hypothesis is that the actin in combination with other substances binds to these specialized membranes and to itself and thus can accumulate in the periacrosmoal region by being trapped on these specialized membranes. Diffusion would then be sufficient to move these substances to this region. In support of this hypothesis are experiments in which I treated mature sperm with detergents, glycols, and hypotonic media, which solubilize or lift away the plasma membrane. The actin and its associated proteins remain attached to these specialized membranes. Thus actin can be nonrandomly distributed in cells in a nonfilamentous state presumably by its association with specialized membranes.


2005 ◽  
Vol 288 (1) ◽  
pp. C46-C56 ◽  
Author(s):  
Camille Ehre ◽  
Andrea H. Rossi ◽  
Lubna H. Abdullah ◽  
Kathleen De Pestel ◽  
Sandra Hill ◽  
...  

Airway goblet cells secrete mucin onto mucosal surfaces under the regulation of an apical, phospholipase C/Gq-coupled P2Y2receptor. We tested whether cortical actin filaments negatively regulate exocytosis in goblet cells by forming a barrier between secretory granules and plasma membrane docking sites as postulated for other secretory cells. Immunostaining of human lung tissues and SPOC1 cells (an epithelial, mucin-secreting cell line) revealed an apical distribution of β- and γ-actin in ciliated and goblet cells. In goblet cells, actin appeared as a prominent subplasmalemmal sheet lying between granules and the apical membrane, and it disappeared from SPOC1 cells activated by purinergic agonist. Disruption of actin filaments with latrunculin A stimulated SPOC1 cell mucin secretion under basal and agonist-activated conditions, whereas stabilization with jasplakinolide or overexpression of β- or γ-actin conjugated to yellow fluorescent protein (YFP) inhibited secretion. Myristoylated alanine-rich C kinase substrate, a PKC-activated actin-plasma membrane tethering protein, was phosphorylated after agonist stimulation, suggesting a translocation to the cytosol. Scinderin (or adseverin), a Ca2+-activated actin filament severing and capping protein was cloned from human airway and SPOC1 cells, and synthetic peptides corresponding to its actin-binding domains inhibited mucin secretion. We conclude that actin filaments negatively regulate mucin secretion basally in airway goblet cells and are dynamically remodeled in agonist-stimulated cells to promote exocytosis.


1988 ◽  
Vol 8 (11) ◽  
pp. 4716-4720
Author(s):  
A J Fornace ◽  
H Schalch ◽  
I Alamo

Sequence analysis of Chinese hamster V79 lung fibroblast cDNA clones, which code for UV radiation-inducible transcripts, revealed that many of the clones corresponded to metallothioneins (MTs) I and II. A third cDNA clone, DDIU4, was found also to code for a similar-size UV-inducible transcript which was unrelated to MT by both sequence analysis and kinetics of induction. MTI and MTII RNAs rapidly increased in V79 cells within 1 h after UV irradiation, and maximum induction was seen by 4 h. This rapid induction of MT RNA by UV irradiation was not observed in human fibroblasts. MTI and MTII were coordinately induced in both time course and dose-response experiments, although the induction of MTII, up to 30-fold, was three to four times greater than that of MTI. The induction of MT did not appear to be a general stress response, since no increase occurred after exposure to X rays or H2O2.


1994 ◽  
Vol 107 (7) ◽  
pp. 1921-1928 ◽  
Author(s):  
K. Takeuchi ◽  
A. Kawashima ◽  
A. Nagafuchi ◽  
S. Tsukita

Several proteins contain the domain homologous to the N-terminal half of band 4.1 protein, indicating the existence of a superfamily. The members of this ‘band 4.1’ superfamily are thought to play crucial roles in the regulation of cytoskeleton-plasma membrane interaction just beneath plasma membranes. We examined the structural diversity of this superfamily by means of the polymerase chain reaction using synthesized mixed primers. We thus identified many members of the band 4.1 superfamily that were expressed in mouse teratocarcinoma F9 cells and mouse brain tissue. In total, 15 cDNA clones were obtained; 8 were identical to the corresponding parts of cDNAs for the known members, while 7 appeared to encode novel proteins (NBL1-7: novel band 4.1-like proteins). Sequence analyses of these clones revealed that the band 4.1 superfamily can be subdivided into 5 gene families; band 4.1 protein, ERM (ezrin/radixin/moesin/merlin/NBL6/NBL7+ ++), talin, PTPH1 (PTPH1/PTPMEG/NBL1-3), and NBL4 (NBL4/NBL5) families. The NBL4 family was first identified here, and the full-length cDNA encoding NBL4 was cloned. The deduced amino acid sequence revealed a myristoylation site, as well as phosphorylation sites for A-kinase and tyrosine kinases in its N-terminal half, suggesting its involvement in the phosphorylation-dependent regulation of cellular events just beneath the plasma membrane. In this study, we describe the initial characterization of these new members and discuss the evolution of the band 4.1 superfamily.


1994 ◽  
Vol 107 (6) ◽  
pp. 1623-1631 ◽  
Author(s):  
M. Footer ◽  
A. Bretscher

The isolated intestinal microvillus cytoskeleton (core) consists of four major proteins: actin, villin, fimbrin and brush border myosin-I. These proteins can assemble in vitro into structures resembling native microvillus cores. Of these components, villin and brush border myosin-I show tissue-specific expression, so they may be involved in the morphogenesis of intestinal microvilli. When introduced into cultured cells that normally lack the protein, villin induces a reorganization of the actin filaments to generate large surface microvilli. Here we examine the consequences of microinjecting brush border myosin-I either alone or together with villin into cultured fibroblasts. Injection of brush border myosin-I has no discernible effect on the overall morphology of the cells, but does become localized to either normal or villin-induced microvilli and other surface structures containing an actin cytoskeleton. Since some endogenous myosin-Is have been found associated with cytoplasmic vesicles, these results show that brush border myosin-I has a domain that specifically targets it to the plasma membrane in both intestinal and cultured cell systems. Ultrastructural examination of microvilli on control cultured cells revealed that they contain a far more highly ordered bundle of microfilaments than had been previously appreciated. The actin filaments in microvilli of villin-injected cells appeared to be more tightly cross-linked when examined by thin-section electron microscopy. In intestinal microvilli, the core bundle is separated from the plasma membrane by about 30 nm due to the presence of brush border myosin-I.(ABSTRACT TRUNCATED AT 250 WORDS)


Sign in / Sign up

Export Citation Format

Share Document