Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Skelet Muscle ; 12(1): 2, 2022 01 22.
Article in English | MEDLINE | ID: mdl-35065666

ABSTRACT

BACKGROUND: The sarcoglycan complex (SC) is part of a network that links the striated muscle cytoskeleton to the basal lamina across the sarcolemma. The SC coordinates changes in phosphorylation and Ca++-flux during mechanical deformation, and these processes are disrupted with loss-of-function mutations in gamma-sarcoglycan (Sgcg) that cause Limb girdle muscular dystrophy 2C/R5. METHODS: To gain insight into how the SC mediates mechano-signaling in muscle, we utilized LC-MS/MS proteomics of SC-associated proteins in immunoprecipitates from enriched sarcolemmal fractions. Criteria for inclusion were co-immunoprecipitation with anti-Sgcg from C57BL/6 control muscle and under-representation in parallel experiments with Sgcg-null muscle and with non-specific IgG. Validation of interaction was performed in co-expression experiments in human RH30 rhabdomyosarcoma cells. RESULTS: We identified 19 candidates as direct or indirect interactors for Sgcg, including the other 3 SC proteins. Novel potential interactors included protein-phosphatase-1-catalytic-subunit-beta (Ppp1cb, PP1b) and Na+-K+-Cl--co-transporter NKCC1 (SLC12A2). NKCC1 co-localized with Sgcg after co-expression in human RH30 rhabdomyosarcoma cells, and its cytosolic domains depleted Sgcg from cell lysates upon immunoprecipitation and co-localized with Sgcg after detergent permeabilization. NKCC1 localized in proximity to the dystrophin complex at costameres in vivo. Bumetanide inhibition of NKCC1 cotransporter activity in isolated muscles reduced SC-dependent, strain-induced increases in phosphorylation of extracellular signal-regulated kinases 1 and 2 (ERK1/2). In silico analysis suggests that candidate SC interactors may cross-talk with survival signaling pathways, including p53, estrogen receptor, and TRIM25. CONCLUSIONS: Results support that NKCC1 is a new SC-associated signaling protein. Moreover, the identities of other candidate SC interactors suggest ways by which the SC and NKCC1, along with other Sgcg interactors such as the membrane-cytoskeleton linker archvillin, may regulate kinase- and Ca++-mediated survival signaling in skeletal muscle.


Subject(s)
Rhabdomyosarcoma , Sarcoglycans , Animals , Chromatography, Liquid , Humans , Mice , Muscle, Skeletal/metabolism , Rhabdomyosarcoma/metabolism , Sarcoglycans/genetics , Solute Carrier Family 12, Member 2/metabolism , Tandem Mass Spectrometry
2.
Brain ; 143(8): 2406-2420, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32779703

ABSTRACT

The muscle specific isoform of the supervillin protein (SV2), encoded by the SVIL gene, is a large sarcolemmal myosin II- and F-actin-binding protein. Supervillin (SV2) binds and co-localizes with costameric dystrophin and binds nebulin, potentially attaching the sarcolemma to myofibrillar Z-lines. Despite its important role in muscle cell physiology suggested by various in vitro studies, there are so far no reports of any human disease caused by SVIL mutations. We here report four patients from two unrelated, consanguineous families with a childhood/adolescence onset of a myopathy associated with homozygous loss-of-function mutations in SVIL. Wide neck, anteverted shoulders and prominent trapezius muscles together with variable contractures were characteristic features. All patients showed increased levels of serum creatine kinase but no or minor muscle weakness. Mild cardiac manifestations were observed. Muscle biopsies showed complete loss of large supervillin isoforms in muscle fibres by western blot and immunohistochemical analyses. Light and electron microscopic investigations revealed a structural myopathy with numerous lobulated muscle fibres and considerable myofibrillar alterations with a coarse and irregular intermyofibrillar network. Autophagic vacuoles, as well as frequent and extensive deposits of lipoproteins, including immature lipofuscin, were observed. Several sarcolemma-associated proteins, including dystrophin and sarcoglycans, were partially mis-localized. The results demonstrate the importance of the supervillin (SV2) protein for the structural integrity of muscle fibres in humans and show that recessive loss-of-function mutations in SVIL cause a distinctive and novel myopathy.


Subject(s)
Membrane Proteins/genetics , Microfilament Proteins/genetics , Muscular Diseases/genetics , Muscular Diseases/pathology , Adolescent , Age of Onset , Autophagy , Child , Female , Humans , Loss of Function Mutation , Male , Muscle, Skeletal/pathology , Pedigree , Vacuoles/pathology
3.
J Biol Chem ; 294(18): 7219-7220, 2019 05 03.
Article in English | MEDLINE | ID: mdl-31053679

ABSTRACT

"Myosin" is famous as a component of muscle fibrils, but the majority of myosin family members act elsewhere with roles unrelated to muscle contraction. The biological functions of a relatively new family of these unconventional myosins, myosins 18A and 18B, are poorly understood. New research from Horsthemke et al. describes a new isoform (Myo18Aγ) that is essential for heart function and viability in mice. Their findings both support and contradict other work in the field and raise new questions about the roles of myosin 18 proteins in vivo.


Subject(s)
Myofibrils/metabolism , Myosins/metabolism , Animals , Cytoskeleton/metabolism , Mice
4.
PLoS One ; 13(10): e0205910, 2018.
Article in English | MEDLINE | ID: mdl-30332471

ABSTRACT

Supervillin isoforms have been implicated in cell proliferation, actin filament-based motile processes, vesicle trafficking, and signal transduction. However, an understanding of the roles of these proteins in cancer metastasis and physiological processes has been limited by the difficulty of obtaining specific antibodies against these highly conserved membrane-associated proteins. To facilitate research into the biological functions of supervillin, monoclonal antibodies were generated against the bacterially expressed human supervillin N-terminus. Two chimeric monoclonal antibodies with rabbit Fc domains (clones 1E2/CPTC-SVIL-1; 4A8/CPTC-SVIL-2) and two mouse monoclonal antibodies (clones 5A8/CPTC-SVIL-3; 5G3/CPTC-SVIL-4) were characterized with respect to their binding sites, affinities, and for efficacy in immunoblotting, immunoprecipitation, immunofluorescence microscopy and immunohistochemical staining. Two antibodies (1E2, 5G3) recognize a sequence found only in primate supervillins, whereas the other two antibodies (4A8, 5A8) are specific for a more broadly conserved conformational epitope(s). All antibodies function in immunoblotting, immunoprecipitation and in immunofluorescence microscopy under the fixation conditions identified here. We also show that the 5A8 antibody works on immunohistological sections. These antibodies should provide useful tools for the study of mammalian supervillins.


Subject(s)
Antibodies, Monoclonal/immunology , Membrane Proteins/immunology , Microfilament Proteins/immunology , Amino Acid Sequence , Animals , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , HeLa Cells , Humans , Kinetics , Membrane Proteins/chemistry , Mice , Microfilament Proteins/chemistry , Muscles/metabolism , Protein Isoforms/immunology , Rats
5.
PLoS One ; 11(7): e0158349, 2016.
Article in English | MEDLINE | ID: mdl-27415442

ABSTRACT

The organ of Corti has evolved a panoply of cells with extraordinary morphological specializations to harness, direct, and transduce mechanical energy into electrical signals. Among the cells with prominent apical specializations are hair cells and nearby supporting cells. At the apical surface of each hair cell is a mechanosensitive hair bundle of filamentous actin (F-actin)-based stereocilia, which insert rootlets into the F-actin meshwork of the underlying cuticular plate, a rigid organelle considered to hold the stereocilia in place. Little is known about the protein composition and development of the cuticular plate or the apicolateral specializations of organ of Corti supporting cells. We show that supervillin, an F-actin cross-linking protein, localizes to cuticular plates in hair cells of the mouse cochlea and vestibule and zebrafish sensory epithelia. Moreover, supervillin localizes near the apicolateral margins within the head plates of Deiters' cells and outer pillar cells, and proximal to the apicolateral margins of inner phalangeal cells, adjacent to the junctions with neighboring hair cells. Overall, supervillin localization suggests this protein may shape the surface structure of the organ of Corti.


Subject(s)
Hair Cells, Auditory/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Organ of Corti/metabolism , Stereocilia/metabolism , Vestibule, Labyrinth/metabolism , Actin Cytoskeleton/metabolism , Animals , Mice , Zebrafish
6.
Cytoskeleton (Hoboken) ; 72(1): 47-64, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25655724

ABSTRACT

We investigated cross-talk between the membrane-associated, myosin II-regulatory protein supervillin and the actin-regulatory small GTPases Rac1, RhoA, and Cdc42. Supervillin knockdown reduced Rac1-GTP loading, but not the GTP loading of RhoA or Cdc42, in HeLa cells with normal levels of the Rac1-activating protein Trio. No reduction in Rac1-GTP loading was observed when supervillin levels were reduced in Trio-depleted cells. Conversely, overexpression of supervillin isoform 1 (SV1) or, especially, isoform 4 (SV4) increased Rac1 activation. Inhibition of the Trio-mediated Rac1 guanine nucleotide exchange activity with ITX3 partially blocked the SV4-mediated increase in Rac1-GTP. Both SV4 and SV1 co-localized with Trio at or near the plasma membrane in ruffles and cell surface projections. Two sequences within supervillin bound directly to Trio spectrin repeats 4-7: SV1-171, which contains N-terminal residues found in both SV1 and SV4 and the SV4-specific differentially spliced coding exons 3, 4, and 5 within SV4 (SV4-E345; SV4 amino acids 276-669). In addition, SV4-E345 interacted with the homologous sequence in rat kalirin (repeats 4-7, amino acids 531-1101). Overexpressed SV1-174 and SV4-E345 affected Rac1-GTP loading, but only in cells with endogenous levels of Trio. Trio residues 771-1057, which contain both supervillin-interaction sites, exerted a dominant-negative effect on cell spreading. Supervillin and Trio knockdowns, separately or together, inhibited cell spreading, suggesting that supervillin regulates the Rac1 guanine nucleotide exchange activity of Trio, and potentially also kalirin, during cell spreading and lamellipodia extension.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , HeLa Cells , Humans , Transfection
7.
Hum Mol Genet ; 24(9): 2470-81, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25605665

ABSTRACT

Loss of gamma-sarcoglycan (γ-SG) induces muscle degeneration and signaling defects in response to mechanical load, and its absence is common to both Duchenne and limb girdle muscular dystrophies. Growing evidence suggests that aberrant signaling contributes to the disease pathology; however, the mechanisms of γ-SG-mediated mechanical signaling are poorly understood. To uncover γ-SG signaling pathway components, we performed yeast two-hybrid screens and identified the muscle-specific protein archvillin as a γ-SG and dystrophin interacting protein. Archvillin protein and message levels were significantly upregulated at the sarcolemma of murine γ-SG-null (gsg(-/-)) muscle but delocalized in dystrophin-deficient mdx muscle. Similar elevation of archvillin protein was observed in human quadriceps muscle lacking γ-SG. Reintroduction of γ-SG in gsg(-/-) muscle by rAAV injection restored archvillin levels to that of control C57 muscle. In situ eccentric contraction of tibialis anterior (TA) muscles from C57 mice caused ERK1/2 phosphorylation, nuclear activation of P-ERK1/2 and stimulus-dependent archvillin association with P-ERK1/2. In contrast, TA muscles from gsg(-/-) and mdx mice exhibited heightened P-ERK1/2 and increased nuclear P-ERK1/2 localization following eccentric contractions, but the archvillin-P-ERK1/2 association was completely ablated. These results position archvillin as a mechanically sensitive component of the dystrophin complex and demonstrate that signaling defects caused by loss of γ-SG occur both at the sarcolemma and in the nucleus.


Subject(s)
Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Muscle, Skeletal/physiology , Sarcoglycans/metabolism , Stress, Mechanical , Animals , Carrier Proteins/metabolism , Cytoskeletal Proteins/metabolism , Dystrophin/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred mdx , Mice, Knockout , Microfilament Proteins/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/metabolism , Muscular Dystrophies, Limb-Girdle/pathology , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Sarcoglycans/chemistry , Sarcoglycans/genetics , Two-Hybrid System Techniques
8.
Am J Pathol ; 184(6): 1831-42, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24726641

ABSTRACT

X-linked myotubular myopathy is a congenital myopathy caused by deficiency of myotubularin. Patients often present with severe perinatal weakness, requiring mechanical ventilation to prevent death from respiratory failure. We recently reported that an activin receptor type IIB inhibitor produced hypertrophy of type 2b myofibers and modest increases of strength and life span in the severely myopathic Mtm1δ4 mouse model of X-linked myotubular myopathy. We have now performed a similar study in the less severely symptomatic Mtm1 p.R69C mouse in hopes of finding greater treatment efficacy. Activin receptor type IIB inhibitor treatment of Mtm1 p.R69C animals produced behavioral and histological evidence of hypertrophy in gastrocnemius muscles but not in quadriceps or triceps. The ability of the muscles to respond to activin receptor type IIB inhibitor treatment correlated with treatment-induced increases in satellite cell number and several muscle-specific abnormalities of hypertrophic signaling. Treatment-responsive Mtm1 p.R69C gastrocnemius muscles displayed lower levels of phosphorylated ribosomal protein S6 and higher levels of phosphorylated eukaryotic elongation factor 2 kinase than were observed in Mtm1 p.R69C quadriceps muscle or in muscles from wild-type littermates. Hypertrophy in the Mtm1 p.R69C gastrocnemius muscle was associated with increased levels of phosphorylated ribosomal protein S6. Our findings indicate that muscle-, fiber type-, and mutation-specific factors affect the response to hypertrophic therapies that will be important to assess in future therapeutic trials.


Subject(s)
Activin Receptors, Type II/metabolism , Muscle Proteins/metabolism , Myopathies, Structural, Congenital/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Signal Transduction , Activin Receptors, Type II/genetics , Animals , Disease Models, Animal , Mice , Mice, Mutant Strains , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/pathology , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Proto-Oncogene Proteins c-akt/genetics , Satellite Cells, Skeletal Muscle/pathology
9.
Mol Biol Cell ; 24(23): 3603-19, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24088567

ABSTRACT

Cytokinesis, the process by which cytoplasm is apportioned between dividing daughter cells, requires coordination of myosin II function, membrane trafficking, and central spindle organization. Most known regulators act during late cytokinesis; a few, including the myosin II-binding proteins anillin and supervillin, act earlier. Anillin's role in scaffolding the membrane cortex with the central spindle is well established, but the mechanism of supervillin action is relatively uncharacterized. We show here that two regions within supervillin affect cell division: residues 831-1281, which bind central spindle proteins, and residues 1-170, which bind the myosin II heavy chain (MHC) and the long form of myosin light-chain kinase. MHC binding is required to rescue supervillin deficiency, and mutagenesis of this site creates a dominant-negative phenotype. Supervillin concentrates activated and total myosin II at the furrow, and simultaneous knockdown of supervillin and anillin additively increases cell division failure. Knockdown of either protein causes mislocalization of the other, and endogenous anillin increases upon supervillin knockdown. Proteomic identification of interaction partners recovered using a high-affinity green fluorescent protein nanobody suggests that supervillin and anillin regulate the myosin II and actin cortical cytoskeletons through separate pathways. We conclude that supervillin and anillin play complementary roles during vertebrate cytokinesis.


Subject(s)
Contractile Proteins/metabolism , Cytokinesis , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Myosin Type II/metabolism , Amino Acid Sequence , Binding Sites , Cytoskeleton/metabolism , Gene Knockdown Techniques , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Membrane Proteins/chemistry , Microfilament Proteins/chemistry , Mutagenesis , Myosin-Light-Chain Kinase/metabolism , Point Mutation , Protein Binding , Protein Transport , Sequence Deletion , Signal Transduction
10.
J Biol Chem ; 288(11): 7918-7929, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23382381

ABSTRACT

Integrin-based adhesions promote cell survival as well as cell motility and invasion. We show here that the adhesion regulatory protein supervillin increases cell survival by decreasing levels of the tumor suppressor protein p53 and downstream target genes. RNAi-mediated knockdown of a new splice form of supervillin (isoform 4) or both isoforms 1 and 4 increases the amount of p53 and cell death, whereas p53 levels decrease after overexpression of either supervillin isoform. Cellular responses to DNA damage induced by etoposide or doxorubicin include down-regulation of endogenous supervillin coincident with increases in p53. In DNA-damaged supervillin knockdown cells, p53 knockdown or inhibition partially rescues the loss of cell metabolic activity, a measure of cell proliferation. Knockdown of the p53 deubiquitinating enzyme USP7/HAUSP also reverses the supervillin phenotype, blocking the increase in p53 levels seen after supervillin knockdown and accentuating the decrease in p53 levels triggered by supervillin overexpression. Conversely, supervillin overexpression decreases the association of USP7 and p53 and attenuates USP7-mediated p53 deubiquitination. USP7 binds directly to the supervillin N terminus and can deubiquitinate and stabilize supervillin. Supervillin also is stabilized by derivatization with the ubiquitin-like protein SUMO1. These results show that supervillin regulates cell survival through control of p53 levels and suggest that supervillin and its interaction partners at sites of cell-substrate adhesion constitute a locus for cross-talk between survival signaling and cell motility pathways.


Subject(s)
Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cloning, Molecular , Cytoskeleton/metabolism , DNA Damage , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Genetic Vectors , HEK293 Cells , HeLa Cells , Humans , Membrane Proteins/chemistry , Microfilament Proteins/chemistry , Microscopy, Fluorescence/methods , Models, Biological , Protein Isoforms , Signal Transduction , Ubiquitin/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Specific Peptidase 7
11.
J Biomol Struct Dyn ; 31(10): 1150-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23075227

ABSTRACT

Supervillin, the largest member of the villin/gelsolin family, is a cytoskeleton regulating, peripheral membrane protein. Supervillin increases cell motility and promotes invasive activity in tumors. Major cytoskeletal interactors, including filamentous actin and myosin II, bind within the unique supervillin amino terminus, amino acids 1-830. The structural features of this key region of the supervillin polypeptide are unknown. Here, we utilize circular dichroism and bioinformatics sequence analysis to demonstrate that the N-terminal part of supervillin forms an extended intrinsically disordered region (IDR). Our combined data indicate that the N-terminus of human and bovine supervillin sequences (positions 1-830) represents an IDR, which is the largest IDR known to date in the villin/gelsolin family. Moreover, this result suggests a potentially novel mechanism of regulation of myosin II and F-actin via the intrinsically disordered N-terminal region of hub protein supervillin.


Subject(s)
Actins/chemistry , Binding Sites , Microfilament Proteins/chemistry , Myosin Type II/chemistry , Protein Interaction Domains and Motifs , Actins/metabolism , Amino Acid Sequence , Amino Acids/chemistry , Animals , Cattle , Chickens , Humans , Microfilament Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Myosin Type II/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Conformation , Sequence Alignment
12.
Circulation ; 125(22): 2762-71, 2012 Jun 05.
Article in English | MEDLINE | ID: mdl-22550155

ABSTRACT

BACKGROUND: High shear force critically regulates platelet adhesion and thrombus formation during ischemic vascular events. To identify genetic factors that influence platelet thrombus formation under high shear stress, we performed a genome-wide association study and confirmatory experiments in human and animal platelets. METHODS AND RESULTS: Closure times in the shear-dependent platelet function analyzer (PFA)-100 were measured on healthy, nondiabetic European Americans (n=125) and blacks (n=116). A genome-wide association (P<5×10(-8)) was identified with 2 single-nucleotide polymorphisms within the SVIL gene (chromosome 10p11.23) in African Americans but not European Americans. Microarray analyses of human platelet RNA demonstrated the presence of SVIL isoform 1 (supervillin) but not muscle-specific isoforms 2 and 3 (archvillin, SmAV). SVIL mRNA levels were associated with SVIL genotypes (P≤0.02) and were inversely correlated with PFA-100 closure times (P<0.04) and platelet volume (P<0.02). Leukocyte-depleted platelets contained abundant levels of the ≈205-kDa supervillin polypeptide. To assess functionality, mice lacking platelet supervillin were generated and back-crossed onto a C57BL/6 background. Compared with controls, murine platelets lacking supervillin were larger by flow cytometry and confocal microscopy and exhibited enhanced platelet thrombus formation under high-shear but not low-shear conditions. CONCLUSIONS: We show for the first time that (1) platelets contain supervillin; (2) platelet thrombus formation in the PFA-100 is associated with human SVIL variants and low SVIL expression; and (3) murine platelets lacking supervillin exhibit enhanced platelet thrombus formation at high shear stress. These data are consistent with an inhibitory role for supervillin in platelet adhesion and arterial thrombosis.


Subject(s)
Blood Platelets/physiology , Genome-Wide Association Study , Membrane Proteins/physiology , Microfilament Proteins/physiology , Platelet Adhesiveness/physiology , Stress, Mechanical , Thrombosis/physiopathology , Adult , Black or African American/genetics , Animals , Blood Platelets/cytology , Cell Size , Female , Genotype , Humans , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Middle Aged , Models, Animal , Polymorphism, Single Nucleotide/genetics , White People/genetics
13.
J Cell Sci ; 125(Pt 9): 2300-14, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22344260

ABSTRACT

Podosomes are actin-rich adhesion and invasion structures. Especially in macrophages, podosomes exist in two subpopulations, large precursors at the cell periphery and smaller podosomes (successors) in the cell interior. To date, the mechanisms that differentially regulate these subpopulations are largely unknown. Here, we show that the membrane-associated protein supervillin localizes preferentially to successor podosomes and becomes enriched at precursors immediately before their dissolution. Consistently, podosome numbers are inversely correlated with supervillin protein levels. Using deletion constructs, we find that the myosin II regulatory N-terminus of supervillin [SV(1-174)] is crucial for these effects. Phosphorylated myosin light chain (pMLC) localizes at supervillin-positive podosomes, and time-lapse analyses show that enrichment of GFP-supervillin at podosomes coincides with their coupling to contractile myosin-IIA-positive cables. We also show that supervillin binds only to activated myosin IIA, and a dysregulated N-terminal construct [SV(1-830)] enhances pMLC levels at podosomes. Thus, preferential recruitment of supervillin to podosome subpopulations might both require and induce actomyosin contractility. Using siRNA and pharmacological inhibition, we demonstrate that supervillin and myosin IIA cooperate to regulate podosome lifetime, podosomal matrix degradation and cell polarization. In sum, we show here that podosome subpopulations differ in their molecular composition and identify supervillin, in cooperation with myosin IIA, as a crucial factor in the regulation of podosome turnover and function.


Subject(s)
Cell Surface Extensions/ultrastructure , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Monocytes/cytology , Nonmuscle Myosin Type IIA/metabolism , Actins/metabolism , Actomyosin/metabolism , Cell Polarity/physiology , Cell Surface Extensions/physiology , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Monocytes/physiology , Monocytes/ultrastructure , Myosin Light Chains/genetics , Myosin Light Chains/metabolism , Nonmuscle Myosin Type IIA/genetics , Phosphorylation , Plasmids , Protein Binding , Protein Structure, Tertiary , RNA, Small Interfering , Time-Lapse Imaging , Transfection
14.
Cytoskeleton (Hoboken) ; 67(6): 346-64, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20309963

ABSTRACT

Supervillin, the largest member of the villin/gelsolin/flightless family, is a peripheral membrane protein that regulates each step of cell motility, including cell spreading. Most known interactors bind within its amino (N)-terminus. We show here that the supervillin carboxy (C)-terminus can be modeled as supervillin-specific loops extending from gelsolin-like repeats plus a villin-like headpiece. We have identified 27 new candidate interactors from yeast two-hybrid screens. The interacting sequences from 12 of these proteins (BUB1, EPLIN/LIMA1, FLNA, HAX1, KIF14, KIFC3, MIF4GD/SLIP1, ODF2/Cenexin, RHAMM, STARD9/KIF16A, Tks5/SH3PXD2A, TNFAIP1) co-localize with and mis-localize EGFP-supervillin in mammalian cells, suggesting associations in vivo. Supervillin-interacting sequences within BUB1, FLNA, HAX1, and MIF4GD also mimic supervillin over-expression by inhibiting cell spreading. Most new interactors have known roles in supervillin-associated processes, e.g. cell motility, membrane trafficking, ERK signaling, and matrix invasion; three (KIF14, KIFC3, STARD9/KIF16A) have kinesin motor domains; and five (EPLIN, KIF14, BUB1, ODF2/cenexin, RHAMM) are important for cell division. GST fusions of the supervillin G2-G3 or G4-G6 repeats co-sediment KIF14 and EPLIN, respectively, consistent with a direct association. Supervillin depletion leads to increased numbers of bi- and multi-nucleated cells. Cytokinesis failure occurs predominately during early cytokinesis. Supervillin localizes with endogenous myosin II and EPLIN in the cleavage furrow, and overlaps with the oncogenic kinesin, KIF14, at the midbody. We conclude that supervillin, like its interactors, is important for efficient cytokinesis. Our results also suggest that supervillin and its interaction partners coordinate actin and microtubule motor functions throughout the cell cycle.


Subject(s)
Cytokinesis , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Amino Acid Sequence , Animals , Biological Assay , COS Cells , Cattle , Cell Proliferation , Chlorocebus aethiops , Cytoskeletal Proteins/metabolism , Gene Knockdown Techniques , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Membrane Proteins/chemistry , Microfilament Proteins/chemistry , Myosin Type II/metabolism , Protein Binding , Protein Structure, Secondary , Protein Transport , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism
15.
Traffic ; 11(6): 782-99, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20331534

ABSTRACT

In migrating cells, the cytoskeleton coordinates signal transduction and redistribution of transmembrane proteins, including integrins and growth factor receptors. Supervillin is an F-actin- and myosin II-binding protein that tightly associates with signaling proteins in cholesterol-rich, 'lipid raft' membrane microdomains. We show here that supervillin also can localize with markers for early and sorting endosomes (EE/SE) and with overexpressed components of the Arf6 recycling pathway in the cell periphery. Supervillin tagged with the photoswitchable fluorescent protein, tdEos, moves both into and away from dynamic structures resembling podosomes at the basal cell surface. Rapid integrin recycling from EE/SE is inhibited in supervillin-knockdown cells, but the rates of integrin endocytosis and recycling from the perinuclear recycling center (PNRC) are unchanged. A lack of synergy between supervillin knockdown and the actin filament barbed-end inhibitor, cytochalasin D, suggests that both treatments affect actin-dependent rapid recycling. Supervillin also enhances signaling from the epidermal growth factor receptor (EGFR) to extracellular signal-regulated kinases (ERKs) 1 and 2 and increases the velocity of cell translocation. These results suggest that supervillin, F-actin and associated proteins coordinate a rapid, basolateral membrane recycling pathway that contributes to ERK signaling and actin-based cell motility.


Subject(s)
Actins/chemistry , Cell Movement , Integrins/metabolism , Membrane Proteins/chemistry , Microfilament Proteins/chemistry , Animals , COS Cells , Chlorocebus aethiops , Cytochalasin D/chemistry , Endocytosis , Endosomes/metabolism , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , HeLa Cells , Humans , Membrane Proteins/physiology , Microfilament Proteins/physiology , Signal Transduction
16.
Mol Biol Cell ; 20(3): 948-62, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19109420

ABSTRACT

Tumor cells use actin-rich protrusions called invadopodia to degrade extracellular matrix (ECM) and invade tissues; related structures, termed podosomes, are sites of dynamic ECM interaction. We show here that supervillin (SV), a peripheral membrane protein that binds F-actin and myosin II, reorganizes the actin cytoskeleton and potentiates invadopodial function. Overexpressed SV induces redistribution of lamellipodial cortactin and lamellipodin/RAPH1/PREL1 away from the cell periphery to internal sites and concomitantly increases the numbers of F-actin punctae. Most punctae are highly dynamic and colocalize with the podosome/invadopodial proteins, cortactin, Tks5, and cdc42. Cortactin binds SV sequences in vitro and contributes to the formation of enhanced green fluorescent protein (EGFP)-SV induced punctae. SV localizes to the cores of Src-generated podosomes in COS-7 cells and with invadopodia in MDA-MB-231 cells. EGFP-SV overexpression increases average numbers of ECM holes per cell; RNA interference-mediated knockdown of SV decreases these numbers. Although SV knockdown alone has no effect, simultaneous down-regulation of SV and the closely related protein gelsolin reduces invasion through ECM. Together, our results show that SV is a component of podosomes and invadopodia and that SV plays a role in invadopodial function, perhaps as a mediator of cortactin localization, activation state, and/or dynamics of metalloproteinases at the ventral cell surface.


Subject(s)
Actins/metabolism , Cytoskeleton/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Pseudopodia/metabolism , Animals , Biomarkers/metabolism , COS Cells , Cattle , Cell Line, Tumor , Chlorocebus aethiops , Cortactin/metabolism , Extracellular Matrix/metabolism , Focal Adhesions/metabolism , Green Fluorescent Proteins/metabolism , Humans , Protein Binding , Protein Transport , Recombinant Fusion Proteins/metabolism , src-Family Kinases/metabolism
17.
J Cell Sci ; 120(Pt 21): 3792-803, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17925381

ABSTRACT

During cell migration, myosin II modulates adhesion, cell protrusion and actin organization at the leading edge. We show that an F-actin- and membrane-associated scaffolding protein, called supervillin (SV, p205), binds directly to the subfragment 2 domains of nonmuscle myosin IIA and myosin IIB and to the N-terminus of the long form of myosin light chain kinase (L-MLCK). SV inhibits cell spreading via an MLCK- and myosin II-dependent mechanism. Overexpression of SV reduces the rate of cell spreading, and RNAi-mediated knockdown of endogenous SV increases it. Endogenous and EGFP-tagged SV colocalize with, and enhance the formation of, cortical bundles of F-actin and activated myosin II during early cell spreading. The effects of SV are reversed by inhibition of myosin heavy chain (MHC) ATPase (blebbistatin), MLCK (ML-7) or MEK (U0126), but not by inhibiting Rho-kinase with Y-27632. Flag-tagged L-MLCK co-localizes in cortical bundles with EGFP-SV, and kinase-dead L-MLCK disorganizes these bundles. The L-MLCK- and myosin-binding site in SV, SV1-171, rearranges and co-localizes with mono- and di-phosphorylated myosin light chain and with L-MLCK, but not with the short form of MLCK (S-MLCK) or with myosin phosphatase. Thus, the membrane protein SV apparently contributes to myosin II assembly during cell spreading by modulating myosin II regulation by L-MLCK.


Subject(s)
Cell Adhesion/physiology , Cell Movement/physiology , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Myosin Type II/metabolism , Actins/metabolism , Animals , COS Cells , Chlorocebus aethiops , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Membrane Proteins/genetics , Mice , Microfilament Proteins/genetics , Myosin Type II/genetics , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/metabolism , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
18.
J Cell Biol ; 174(3): 447-58, 2006 Jul 31.
Article in English | MEDLINE | ID: mdl-16880273

ABSTRACT

Cell-substrate contacts, called focal adhesions (FAs), are dynamic in rapidly moving cells. We show that supervillin (SV)--a peripheral membrane protein that binds myosin II and F-actin in such cells--negatively regulates stress fibers, FAs, and cell-substrate adhesion. The major FA regulatory sequence within SV (SV342-571) binds to the LIM domains of two proteins in the zyxin family, thyroid receptor-interacting protein 6 (TRIP6) and lipoma-preferred partner (LPP), but not to zyxin itself. SV and TRIP6 colocalize within large FAs, where TRIP6 may help recruit SV. RNAi-mediated decreases in either protein increase cell adhesion to fibronectin. TRIP6 partially rescues SV effects on stress fibers and FAs, apparently by mislocating SV away from FAs. Thus, SV interactions with TRIP6 at FAs promote loss of FA structure and function. SV and TRIP6 binding partners suggest several specific mechanisms through which the SV-TRIP6 interaction may regulate FA maturation and/or disassembly.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Focal Adhesions/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Transcription Factors/metabolism , ATPases Associated with Diverse Cellular Activities , Adaptor Proteins, Signal Transducing/chemistry , Animals , COS Cells , Cattle , Cells, Cultured , Chlorocebus aethiops , Down-Regulation/genetics , Green Fluorescent Proteins/metabolism , Humans , LIM Domain Proteins , Mice , Microtubule-Associated Proteins/metabolism , Myocytes, Smooth Muscle/cytology , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex , Protein Binding , Rats , Regulatory Sequences, Nucleic Acid/genetics , Transcription Factors/chemistry , t-Complex Genome Region
19.
Plant Physiol ; 136(2): 3080-94; discussion 3001-2, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15466235

ABSTRACT

Membrane trafficking is central to construction of the cell plate during plant cytokinesis. Consequently, a detailed understanding of the process depends on the characterization of molecules that function in the formation, transport, targeting, and fusion of membrane vesicles to the developing plate, as well as those that participate in its consolidation and maturation into a fully functional partition. Here we report the initial biochemical and functional characterization of patellin1 (PATL1), a novel cell-plate-associated protein that is related in sequence to proteins involved in membrane trafficking in other eukaryotes. Analysis of the Arabidopsis genome indicated that PATL1 is one of a small family of Arabidopsis proteins, characterized by a variable N-terminal domain followed by two domains found in other membrane-trafficking proteins (Sec14 and Golgi dynamics domains). Results from immunolocalization and biochemical fractionation studies suggested that PATL1 is recruited from the cytoplasm to the expanding and maturing cell plate. In vesicle-binding assays, PATL1 bound to specific phosphoinositides, important regulators of membrane trafficking, with a preference for phosphatidylinositol(5)P, phosphatidylinositol(4,5)P(2), and phosphatidylinositol(3)P. Taken together, these findings suggest a role for PATL1 in membrane-trafficking events associated with cell-plate expansion or maturation and point to the involvement of phosphoinositides in cell-plate biogenesis.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Phosphatidylinositols/metabolism , Phospholipid Transfer Proteins/metabolism , Amino Acid Sequence , Arabidopsis Proteins/chemistry , Gene Expression Regulation, Plant , Microscopy, Fluorescence , Molecular Sequence Data , Multigene Family , Phospholipid Transfer Proteins/chemistry , Plant Roots/metabolism , Protein Binding , Sequence Alignment , Sequence Homology, Amino Acid , Nicotiana/metabolism
20.
J Cell Sci ; 117(Pt 21): 5043-57, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15383618

ABSTRACT

The mechanisms by which protein kinase C (PKC) and extracellular-signal-regulated kinases (ERK1/2) govern smooth-muscle contractility remain unclear. Calponin (CaP), an actin-binding protein and PKC substrate, mediates signaling through ERK1/2. We report here that CaP sequences containing the CaP homology (CH) domain bind to the C-terminal 251 amino acids of smooth-muscle archvillin (SmAV), a new splice variant of supervillin, which is a known actin- and myosin-II-binding protein. The CaP-SmAV interaction is demonstrated by reciprocal yeast two-hybrid and blot-overlay assays and by colocalization in COS-7 cells. In differentiated smooth muscle, endogenous SmAV and CaP co-fractionate and co-translocate to the cell cortex after stimulation by agonist. Antisense knockdown of SmAV in tissue inhibits both the activation of ERK1/2 and contractions stimulated by either agonist or PKC activation. This ERK1/2 signaling and contractile defect is similar to that observed in CaP knockdown experiments. In A7r5 smooth-muscle cells, PKC activation by phorbol esters induces the reorganization of endogenous, membrane-localized SmAV and microfilament-associated CaP into podosome-like structures that also contain F-actin, nonmuscle myosin IIB and ERK1/2. These results indicate that SmAV contributes to the regulation of contractility through a CaP-mediated signaling pathway, involving PKC activation and phosphorylation of ERK1/2.


Subject(s)
Membrane Proteins/physiology , Microfilament Proteins/physiology , Muscle, Smooth/metabolism , Alternative Splicing , Amino Acid Sequence , Animals , Aorta/metabolism , Blotting, Western , COS Cells , Calcium-Binding Proteins/metabolism , DNA, Complementary/metabolism , Enzyme Activation , Ferrets , Glutathione Transferase/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Models, Genetic , Molecular Sequence Data , Oligonucleotides, Antisense/pharmacology , Phosphorylation , Protein Binding , Protein Kinase C/metabolism , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Signal Transduction , Subcellular Fractions/metabolism , Time Factors , Transfection , Two-Hybrid System Techniques , Calponins
SELECTION OF CITATIONS
SEARCH DETAIL
...