Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Oncogene ; 39(12): 2493-2508, 2020 03.
Article in English | MEDLINE | ID: mdl-31996784

ABSTRACT

Early studies indicated that estrogen receptor α (ERα) might impact the progression of hepatocellular carcinoma (HCC). However, the detailed mechanisms, especially its linkage to the gelsolin (GSN)-mediated cell invasion, remain unclear. Here we found that ERα could decrease HCC cell invasion via suppressing the circular RNA-SMG1.72 (circRNA-SMG1.72) expression via transcriptional regulation through directly binding to the 5' promoter region of its host gene SMG1, We showed that ERα-suppressed circ-SMG1.72 could sponge and inhibit the expression of the microRNA (miRNA, miR), miR-141-3p, which could then result in increasing the GSN messenger RNA translation via reduced miR binding to its 3' untranslated region (3'UTR). The preclinical study using an in vivo mouse model with orthotopic xenografts of HCC cells confirmed the in vitro data, and the human HCC clinical sample survey and tissue staining also confirmed the linkage of ERα/miR-141-3p/GSN signaling to the HCC progression. Together, our findings suggest that ERα can suppress HCC cell invasion via altering the ERα/circRNA-SMG1.72/miR-141-3p/GSN signaling, and targeting this newly identified signaling with small molecules may help in the development of novel therapies to better suppress the HCC progression.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Estrogen Receptor alpha/metabolism , Gelsolin/metabolism , Liver Neoplasms/metabolism , MicroRNAs/metabolism , Neoplasm Invasiveness , Protein Serine-Threonine Kinases/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Drug Delivery Systems , Estrogen Receptor alpha/antagonists & inhibitors , Gelsolin/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , Liver Neoplasms/drug therapy , MicroRNAs/antagonists & inhibitors , Neoplasm Invasiveness/prevention & control , RNA, Circular/drug effects , RNA, Neoplasm/drug effects , RNA, Neoplasm/metabolism , Signal Transduction/drug effects , Survival Rate
2.
Antiviral Res ; 143: 252-261, 2017 07.
Article in English | MEDLINE | ID: mdl-28483551

ABSTRACT

Influenza viruses are one of the most important respiratory pathogens worldwide, causing both epidemic and pandemic infections. The aim of the study was to evaluate the effect of FPR2 antagonists PBP10 and BOC2 on influenza virus replication. We determined that these molecules exhibit antiviral effects against influenza A (H1N1, H3N2, H6N2) and B viruses. FPR2 antagonists used in combination with oseltamivir showed additive antiviral effects. Mechanistically, the antiviral effect of PBP10 and BOC2 is mediated through early inhibition of virus-induced ERK activation. Finally, our preclinical studies showed that FPR2 antagonists protected mice from lethal infections induced by influenza, both in a prophylactic and therapeutic manner. Thus, FPR2 antagonists might be explored for novel treatments against influenza.


Subject(s)
Antiviral Agents/pharmacology , Gelsolin/antagonists & inhibitors , Influenza A virus/drug effects , Oligopeptides/antagonists & inhibitors , Peptide Fragments/antagonists & inhibitors , Receptors, Formyl Peptide/antagonists & inhibitors , Receptors, Lipoxin/antagonists & inhibitors , Virus Replication/drug effects , A549 Cells/drug effects , Animals , Antiviral Agents/administration & dosage , Cell Survival , Dogs , Drug Combinations , Female , Gelsolin/administration & dosage , Haplorhini , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/drug effects , Influenza B virus/drug effects , Influenza, Human/drug therapy , Influenza, Human/virology , MAP Kinase Signaling System/drug effects , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred C57BL , Oligopeptides/administration & dosage , Oseltamivir/pharmacology , Peptide Fragments/administration & dosage , Sequence Alignment
3.
Cell Physiol Biochem ; 41(1): 205-212, 2017.
Article in English | MEDLINE | ID: mdl-28135711

ABSTRACT

BACKGROUND/AIMS: Endotoxemia is a life-threatening situation that signifies a key challenge in the field of intensive care medicine. Proinflammatory mediators produced by macrophages play a key role in endotoxemia. Gelsolin (GSN) is involved in the process of inflammation. METHODS: IL-6 and TNF-α in the supernatant were measured with an ELISA kit. NO production was assessed by measurement of nitrite concentration with the Griess assay. si-RNA directed against GSN (si-GSN) was transfected by Lipofectamine. RESULTS: LPS decreased the levels of GSN. Recombinant GSN inhibited the cytokines induced by LPS and rescued mice from LPS-induced death, and si-GSN increased death in the LPS-pretreated mice. CONCLUSION: GSN exhibited a protective role in endotoxemia.


Subject(s)
Endotoxemia/etiology , Gelsolin/metabolism , Lipopolysaccharides/toxicity , Animals , Apoptosis/drug effects , Endotoxemia/mortality , Endotoxemia/pathology , Enzyme-Linked Immunosorbent Assay , Gelsolin/antagonists & inhibitors , Gelsolin/genetics , Interleukin-6/analysis , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , RNA Interference , RNA, Small Interfering/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Tumor Necrosis Factor-alpha/analysis , Up-Regulation/drug effects
4.
Hum Mol Genet ; 24(9): 2492-507, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25601851

ABSTRACT

Hereditary gelsolin amyloidosis is an autosomal dominantly inherited amyloid disorder. A point mutation in the GSN gene (G654A being the most common one) results in disturbed calcium binding by the second gelsolin domain (G2). As a result, the folding of G2 is hampered, rendering the mutant plasma gelsolin susceptible to a proteolytic cascade. Consecutive cleavage by furin and MT1-MMP-like proteases generates 8 and 5 kDa amyloidogenic peptides that cause neurological, ophthalmological and dermatological findings. To this day, no specific treatment is available to counter the pathogenesis. Using GSN nanobody 11 as a molecular chaperone, we aimed to protect mutant plasma gelsolin from furin proteolysis in the trans-Golgi network. We report a transgenic, GSN nanobody 11 secreting mouse that was used for crossbreeding with gelsolin amyloidosis mice. Insertion of the therapeutic nanobody gene into the gelsolin amyloidosis mouse genome resulted in improved muscle contractility. X-ray crystal structure determination of the gelsolin G2:Nb11 complex revealed that Nb11 does not directly block the furin cleavage site. We conclude that nanobodies can be used to shield substrates from aberrant proteolysis and this approach might establish a novel therapeutic strategy in amyloid diseases.


Subject(s)
Amyloid/metabolism , Amyloidosis, Familial/metabolism , Endoplasmic Reticulum/metabolism , Gelsolin/metabolism , Single-Domain Antibodies/pharmacology , Amyloidosis, Familial/genetics , Amyloidosis, Familial/physiopathology , Animals , Disease Models, Animal , Furin/metabolism , Gelsolin/antagonists & inhibitors , Gelsolin/chemistry , Gelsolin/genetics , Gene Expression , HEK293 Cells , Humans , Mice , Mice, Transgenic , Muscle Contraction , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Mutation , Protein Binding , Protein Conformation , Proteolysis/drug effects , Single-Domain Antibodies/chemistry , trans-Golgi Network/metabolism
5.
Int J Oncol ; 44(1): 161-6, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24212916

ABSTRACT

SCIN is a calcium regulated actin severing and capping protein. Its homologue in zebrafish is found to be related with cell death. In the present study, we found that SCIN is highly expressed in human prostate cancer specimens. However, the functions of SCIN in human prostate carcinoma cells are largely unknown. To address the function of SCIN in prostate carcinoma cells, we used lentivirus-mediated RNAi to knock down SCIN expression in PC3 cells, a prostate carcinoma cell line. We found that in vitro silencing of SCIN could inhibit the proliferation and colony formation ability of PC3 cells. Furthermore, cell cycle analysis showed that reduced SCIN expression lead to G0/G1 cell cycle arrest through the regulation of cell cycle-related genes, such as p21Waf1/Cip1, cyclin-dependent kinase inhibitor 2A (CDKN2A, p16Ink4A) and cyclin A2. These results suggest that SCIN plays an important role in the proliferation of prostate cancer cells and lentivirus-mediated inhibition of SCIN expression may be a potential therapeutic method for the treatment of prostate cancer.


Subject(s)
Cell Proliferation , G1 Phase Cell Cycle Checkpoints/genetics , Gelsolin/biosynthesis , Prostatic Neoplasms/genetics , Apoptosis/genetics , Gelsolin/antagonists & inhibitors , Gelsolin/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Lentivirus , Male , Prostatic Neoplasms/pathology
6.
Cancer Res ; 73(19): 5949-62, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23940300

ABSTRACT

Nm23-H1 has been identified as a metastasis suppressor gene, but its protein interactions have yet to be understood with any mechanistic clarity. In this study, we evaluated the proteomic spectrum of interactions made by Nm23-H1 in 4T1 murine breast cancer cells derived from tissue culture, primary mammary tumors, and pulmonary metastases. By this approach, we identified the actin-severing protein Gelsolin as binding partner for Nm23-H1, verifying their interaction by coimmunoprecipitation in 4T1 cells as well as in human MCF7, MDA-MB-231T, and MDA-MB-435 breast cancer cells. In Gelsolin-transfected cells, coexpression of Nm23-H1 abrogated the actin-severing activity of Gelsolin. Conversely, actin severing by Gelsolin was abrogated by RNA interference-mediated silencing of endogenous Nm23-H1. Tumor cell motility was negatively affected in parallel with Gelsolin activity, suggesting that Nm23-H1 binding inactivated the actin-depolymerizing function of Gelsolin to inhibit cell motility. Using indirect immunoflourescence to monitor complexes formed by Gelsolin and Nm23-H1 in living cells, we observed their colocalization in a perinuclear cytoplasmic compartment that was associated with the presence of disrupted actin stress fibers. In vivo analyses revealed that Gelsolin overexpression increased the metastasis of orthotopically implanted 4T1 or tail vein-injected MDA-MB-231T cells (P = 0.001 and 0.04, respectively), along with the proportion of mice with diffuse liver metastases, an effect ablated by coexpression of Nm23-H1. We observed no variation in proliferation among lung metastases. Our findings suggest a new actin-based mechanism that can suppress tumor metastasis.


Subject(s)
Actins/metabolism , Cell Movement , Gelsolin/metabolism , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Mammary Neoplasms, Animal/pathology , NM23 Nucleoside Diphosphate Kinases/metabolism , Animals , Apoptosis , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Female , Fluorescent Antibody Technique , Gelsolin/antagonists & inhibitors , Gelsolin/genetics , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Immunoprecipitation , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , NM23 Nucleoside Diphosphate Kinases/antagonists & inhibitors , NM23 Nucleoside Diphosphate Kinases/genetics , RNA, Small Interfering/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
7.
Cytoskeleton (Hoboken) ; 70(7): 385-93, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23595955

ABSTRACT

Integrins are functionally regulated by "inside-out" signaling, in that stimulus-induced signaling pathways act on the intracellular integrin tail to regulate the activity of the receptor on the outside. Both a change in conformation (affinity) and clustering (avidity/valency) of the receptors occurs, but the mechanisms that regulate inside out signaling are not completely understood. Previously, we identified gelsolin in a proteomics screen to identify proteins involved in inside-out control of integrins using the lymphocytic leukemia cell line L1210. Furthermore, we showed that gelsolin was involved in affinity regulation of ß1 -integrins in the leukemic cell line U937. Here, we examined how gelsolin regulates ß1 -integrin affinity in the leukemia cell line L1210. We show that gelsolin is mainly expressed at the cell membrane and is present near ß1 -integrins. The role for actin polymerization in integrin affinity regulation was examined using the actin modulating agent jasplakinolide, which decreased ß1 -integrin affinity. Similarly, knock-down of gelsolin in L1210 cells also decreased ß1 -integrin affinity and cell adhesion to collagen. These data suggest that increased actin polymerization through gelsolin regulates ß1 -integrin affinity and cell adhesion.


Subject(s)
Cell Adhesion/physiology , Gelsolin/biosynthesis , Integrin beta1/metabolism , Actins/metabolism , Animals , Cell Membrane/metabolism , Gelsolin/antagonists & inhibitors , Gelsolin/deficiency , Gelsolin/metabolism , Gene Knockdown Techniques , Humans , Integrin beta1/biosynthesis , Leukemia L1210 , Mice , Signal Transduction
8.
Immunity ; 36(5): 795-806, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22633459

ABSTRACT

Primary resistance to pathogens is reliant on both basal and inducible immune defenses. To date, research has focused upon inducible innate immune responses. In contrast to resistance via cytokine induction, basal defense mechanisms are less evident. Here we showed that the antiviral protein kinase R (PKR) inhibited the key actin-modifying protein gelsolin to regulate actin dynamics and control cytoskeletal cellular functions under homeostatic conditions. Through this mechanism, PKR controlled fundamental innate immune, actin-dependent processes that included membrane ruffling and particle engulfment. Accordingly, PKR counteracted viral entry into the cell. These findings identify a layer of host resistance, showing that the regulation of actin-modifying proteins during the innate immune response bolsters first-line defense against intracellular pathogens and has a sustained effect on virus production. Moreover, these data provide proof of principle for a concept in which the cell cytoskeleton could be targeted to elicit broad antiviral protection.


Subject(s)
Actins/metabolism , Gelsolin/metabolism , Immunity, Innate/immunology , eIF-2 Kinase/metabolism , Actins/immunology , Cell Line, Transformed , Cell Line, Tumor , Cytokines/immunology , Cytokines/metabolism , Cytoskeleton/immunology , Cytoskeleton/metabolism , Gelsolin/antagonists & inhibitors , Gelsolin/immunology , HEK293 Cells , HeLa Cells , Humans , Microfilament Proteins/immunology , Microfilament Proteins/metabolism , Protein Interaction Domains and Motifs/immunology , Viruses/immunology , Viruses/metabolism , eIF-2 Kinase/immunology
9.
Neurodegener Dis ; 8(5): 375-80, 2011.
Article in English | MEDLINE | ID: mdl-21389683

ABSTRACT

BACKGROUND/AIMS: Cell damage during the course of inflammation results in cytoplasmic actin release, which if not eliminated by the extracellular actin scavenger system, composed of gelsolin and vitamin D binding protein, can cause dysfunction of hemostasis and toxicity towards surrounding cells. In this study, we test the hypothesis that an inflammatory reaction induced by central nervous system infections such as tick-borne encephalitis (TBE) or Lyme neuroborreliosis (LNB) will result in plasma gelsolin concentration changes in the blood and cerebrospinal fluid (CSF). METHODS: Quantitative Western blot was used to determine gelsolin levels in 58 samples, which include: 29 patients without infection (diagnosed with conditions such as idiopathic cephalalgia, idiopathic Bell's facial nerve palsy and ischialgia due to discopathy in which standard CSF diagnostic tests show no abnormalities), 12 patients diagnosed with TBE, and 17 patients diagnosed with LNB sub forma meningitis. RESULTS AND CONCLUSION: The gelsolin concentration in the blood of patients with TBE (163.2 ± 80.8 µg/ml) and LNB (113.6 ± 56.8 µg/ml) was significantly lower (p < 0.05 and p < 0.001, respectively) compared to the control group (226.3 ± 100.7 µg/ml). Furthermore, there was no statistically significant difference between the CSF gelsolin concentration in patients with TBE (3.9 ± 3.3 µg/ml), LNB (2.9 ± 1.2 µg/ml) and the control group (3.7 ± 3.3 µg/ml). An observed decrease in gelsolin concentration in the blood of TBE and LNB patients supports previous findings indicating the involvement of gelsolin in the pathophysiology of an inflammatory response. Therefore, evaluation of blood gelsolin concentration and administration of recombinant plasma gelsolin might provide a new tool to develop diagnostic and therapeutic strategies for TBE and LNB.


Subject(s)
Down-Regulation/physiology , Encephalitis, Tick-Borne/blood , Encephalitis, Tick-Borne/diagnosis , Gelsolin/blood , Lyme Neuroborreliosis/blood , Lyme Neuroborreliosis/diagnosis , Adult , Aged , Biomarkers/blood , Encephalitis, Tick-Borne/physiopathology , Gelsolin/antagonists & inhibitors , Humans , Lyme Neuroborreliosis/physiopathology , Middle Aged
10.
Biochem Biophys Res Commun ; 392(2): 118-23, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-20035726

ABSTRACT

Gelsolin and calponin are cytoskeletal and signalling proteins that form a tight 1:1 complex (GCC). We show that calponin within the GCC inhibits the rate of gelsolin mediated nucleation of actin polymerization. The actin-binding function of calponin is ablated within the GCC as the actin-binding site overlaps with one of the gelsolin binding sites. The structure of filaments that result from nucleation by GCC are different to those nucleated by gelsolin alone in that they are longer, loosely bundled and stain heterogeneously with phalloidin. GCC nucleated filaments appear contorted and wrap around each to form the loose bundles.


Subject(s)
Actin Cytoskeleton/metabolism , Calcium-Binding Proteins/metabolism , Gelsolin/antagonists & inhibitors , Microfilament Proteins/metabolism , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/ultrastructure , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/pharmacology , Gelsolin/chemistry , Gelsolin/metabolism , Humans , Microfilament Proteins/chemistry , Microfilament Proteins/pharmacology , Microscopy, Electron , Calponins
11.
J Immunol ; 181(7): 4936-44, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18802097

ABSTRACT

The various functions of gelsolin in extracellular compartments are not yet clearly defined but include actin scavenging and antiinflammatory effects. Gelsolin was recently reported to bind endotoxin (LPS) from various Gram-negative bacteria with high affinity. In this study we investigate whether gelsolin also interacts with bacterial wall molecules of Gram-positive bacteria such as lipoteichoic acid (LTA) and whether gelsolin's interaction with bacterial lipids from Gram-negative or Gram-positive bacteria affects their cellular inflammatory responses. A peptide based on the PPI binding site of gelsolin (160-169) binds purified LTA at the same molecular ratio that it binds phosphatidylinositol 4,5-bisphosphate. The OD of recombinant human plasma gelsolin was found to decrease following the addition of purified LTA, and the binding of gelsolin to LTA inhibits F-actin depolymerization by gelsolin. Simultaneously, the ability of LTA to activate translocation of NF-kappaB, E-selectin expression, and adhesion of neutrophils to LTA-treated human aortic endothelial cells was compromised by gelsolin. Gelsolin was able to partially inhibit LPS- or LTA-induced release of IL-8 from human neutrophils but was unable to prevent Gram-positive Bacillus subtilis or Gram-negative Pseudomonas aeruginosa growth and had no effect on the antibacterial activity of the cathelicidin-derived antibacterial peptide LL37. These data suggest that extracellular gelsolin is involved in the host immune recognition of LTA or LPS following release of these molecules from the bacterial outer membrane during cell division or attack by drugs and immune components.


Subject(s)
Cell Wall/immunology , Extracellular Fluid/metabolism , Gelsolin/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/metabolism , Neutrophils/pathology , Staphylococcus aureus/immunology , Teichoic Acids/metabolism , Actins/antagonists & inhibitors , Actins/metabolism , Cell Adhesion/immunology , Cell Wall/metabolism , Cell Wall/pathology , Cells, Cultured , E-Selectin/biosynthesis , E-Selectin/genetics , E-Selectin/metabolism , Extracellular Fluid/cytology , Extracellular Fluid/immunology , Gelsolin/antagonists & inhibitors , Gelsolin/chemical synthesis , Humans , Immunity, Cellular , Inflammation Mediators/antagonists & inhibitors , Interleukin-8/antagonists & inhibitors , Interleukin-8/metabolism , Lipopolysaccharides/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Neutrophils/metabolism , Neutrophils/microbiology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemical synthesis , Peptide Fragments/metabolism , Protein Binding/immunology , Staphylococcus aureus/metabolism , Teichoic Acids/antagonists & inhibitors
12.
Biochem J ; 396(3): 461-8, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16536729

ABSTRACT

Gelsolin and calponin are well-characterized cytoskeletal proteins that are abundant and widely expressed in vertebrate tissues. It is also becoming apparent, however, that they are involved in cell signalling. In the present study, we show that gelsolin and calponin interact directly to form a high-affinity (K(d)=16 nM) 1:1 complex, by the use of fluorescent probes attached to both proteins, by affinity chromatography and by immunoprecipitation. These methods show that gelsolin can form high-affinity complexes with two calponin isoforms (basic h1 and acidic h3). They also show that gelsolin binds calponin through regions that have been identified previously as being calponin's actin-binding sites. Moreover, gelsolin does not interact with calponin while calponin is bound to F-actin. Reciprocal experiments to find calponin-binding sites on gelsolin show that these are in both the N- and C-terminal halves of gelsolin. Calponin has minimal effects on actin severing by gelsolin. In contrast, calponin markedly affects the nucleation activity of gelsolin. The maximum inhibition of nucleation by gelsolin was 50%, which was achieved with a ratio of two calponins for every gelsolin. Thus the interaction of calponin with gelsolin may play a regulatory role in the formation of actin filaments through modulation of gelsolin's actin-binding function and through the prevention of calponin's actin-binding activities.


Subject(s)
Actins/metabolism , Calcium-Binding Proteins/chemistry , Gelsolin/chemistry , Microfilament Proteins/chemistry , Animals , COS Cells , Calcium-Binding Proteins/metabolism , Chlorocebus aethiops , Fluorescent Dyes , Gelsolin/antagonists & inhibitors , Humans , Microfilament Proteins/metabolism , Rats , Spectrometry, Fluorescence , Calponins
13.
Leuk Res ; 29(12): 1443-53, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16242777

ABSTRACT

Viscum album agglutinin-I (VAA-I) is a potent inducer of cell apoptosis and possesses anti-tumoral activity. Using PLB-985 and chronic granulomatous disease (X-CGD) cells, which lack expression of gp91(phox), VAA-I was found to induce apoptosis in both cell lines as assessed by cytology, DNA laddering and degradation of the cytoskeletal protein gelsolin. Both cell lines expressed caspase-3 and -8 and VAA-I activated these caspases. We demonstrated that lamin B(1) is a novel target to VAA-I and its degradation was reversed by a pan-caspase inhibitor and by a caspase-6, but not a caspase-8, inhibitor.


Subject(s)
Apoptosis/drug effects , Cytoskeletal Proteins/drug effects , Leukemia/pathology , Plant Lectins/pharmacology , Plant Preparations/pharmacology , Plant Proteins/pharmacology , Toxins, Biological/pharmacology , Caspase 3 , Caspase 8 , Caspases/drug effects , Caspases/metabolism , Cell Line, Tumor , Cytoskeletal Proteins/metabolism , Gelsolin/antagonists & inhibitors , Humans , Lamin Type B/antagonists & inhibitors , Leukemia/drug therapy , Ribosome Inactivating Proteins , Ribosome Inactivating Proteins, Type 2 , Viscum album/chemistry
14.
Biochemistry ; 44(28): 9590-7, 2005 Jul 19.
Article in English | MEDLINE | ID: mdl-16008344

ABSTRACT

Septic shock from bacterial endotoxin, triggered by the release of lipopolysaccharide (LPS) molecules from the outer wall of Gram-negative bacteria, is a major cause of human death for which there is no effective treatment once the complex inflammatory pathways stimulated by these small amphipathic molecules are activated. Here we report that plasma gelsolin, a highly conserved human protein, binds LPS from various bacteria with high affinity. Solid-phase binding assays, fluorescence measurements, and functional assays of actin depolymerizing effects show that gelsolin binds more tightly to LPS than it does to its other known lipid ligands, phosphatidylinositol 4,5-bisphosphate and lysophosphatidic acid. Gelsolin also competes with LPS-binding protein (LBP), a high-affinity carrier for LPS. One result of gelsolin-LPS binding is inhibition of the actin binding activity of gelsolin as well as the actin depolymerizing activity of blood serum. Simultaneously, effects of LPS on cellular functions, including cytoskeletal actin remodeling, and collagen-induced platelet activation by pathways independent of toll-like receptors (TLRs) are neutralized by gelsolin and by a peptide based on gelsolin residues 160-169 (GSN160-169) which comprise part of gelsolin's phosphoinositide binding site. Additionally, TLR-dependent NF-kappaB translocation in astrocytes appears to be blocked by gelsolin. These results show a strong effect of LPS on plasma gelsolin function and suggest that some effects of endotoxin in vivo may be mediated or inhibited by plasma gelsolin.


Subject(s)
Gelsolin/blood , Gelsolin/chemistry , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/metabolism , Actin Cytoskeleton/metabolism , Acute-Phase Proteins/chemistry , Acute-Phase Proteins/metabolism , Animals , Astrocytes/metabolism , Binding, Competitive , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Line , Cells, Cultured , Gelsolin/antagonists & inhibitors , Humans , Lipopolysaccharides/chemistry , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , NF-kappa B/metabolism , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/blood , Peptide Fragments/chemistry , Phosphatidylinositol Phosphates/chemistry , Phosphatidylinositol Phosphates/metabolism , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/metabolism , Protein Binding , Protein Transport , Rabbits , Rats
15.
J Biol Chem ; 279(35): 36680-8, 2004 Aug 27.
Article in English | MEDLINE | ID: mdl-15213223

ABSTRACT

Activation of Ras promotes oncogenesis by altering a multiple of cellular processes, such as cell cycle progression, differentiation, and apoptosis. Oncogenic Ras can either promote or inhibit apoptosis, depending on the cell type and the nature of the apoptotic stimuli. The response of normal and transformed colonic epithelial cells to the short chain fatty acid butyrate, a physiological regulator of epithelial cell maturation, is also divergent: normal epithelial cells proliferate, and transformed cells undergo apoptosis in response to butyrate. To investigate the role of k-ras mutations in butyrate-induced apoptosis, we utilized HCT116 cells, which harbor an oncogenic k-ras mutation and two isogenic clones with targeted inactivation of the mutant k-ras allele, Hkh2, and Hke-3. We demonstrated that the targeted deletion of the mutant k-ras allele is sufficient to protect epithelial cells from butyrate-induced apoptosis. Consistent with this, we showed that apigenin, a dietary flavonoid that has been shown to inhibit Ras signaling and to reverse transformation of cancer cell lines, prevented butyrate-induced apoptosis in HCT116 cells. To investigate the mechanism whereby activated k-ras sensitizes colonic cells to butyrate, we performed a genome-wide analysis of Ras target genes in the isogenic cell lines HCT116, Hkh2, and Hke-3. The gene exhibiting the greatest down-regulation by the activating k-ras mutation was gelsolin, an actin-binding protein whose expression is frequently reduced or absent in colorectal cancer cell lines and primary tumors. We demonstrated that silencing of gelsolin expression by small interfering RNA sensitized cells to butyrate-induced apoptosis through amplification of the activation of caspase-9 and caspase-7. These data therefore demonstrate that gelsolin protects cells from butyrate-induced apoptosis and suggest that Ras promotes apoptosis, at least in part, through its ability to down-regulate the expression of gelsolin.


Subject(s)
Apoptosis , Butyrates/pharmacology , Gelsolin/antagonists & inhibitors , Oncogene Protein p21(ras)/physiology , Alleles , Antineoplastic Agents/pharmacology , Apigenin , Caspase 7 , Caspase 9 , Caspases/metabolism , Cell Division , Cell Line, Tumor , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Enzyme Activation , Epithelial Cells/metabolism , Flavonoids/metabolism , Gelsolin/metabolism , Gene Deletion , Gene Expression Regulation , Gene Silencing , Genes, Reporter , Genes, ras , Humans , Microscopy, Fluorescence , Mutation , Oligonucleotide Array Sequence Analysis , Oncogene Protein p21(ras)/metabolism , RNA/metabolism , RNA, Small Interfering/metabolism , Sulindac/pharmacology , Time Factors , Transcription, Genetic , Transfection
16.
Proc Natl Acad Sci U S A ; 100(6): 3287-92, 2003 Mar 18.
Article in English | MEDLINE | ID: mdl-12629212

ABSTRACT

The gene for gelsolin (an actin-binding, cytoskeletal regulatory protein) was shown earlier to be specialized for high corneal expression in adult zebrafish. We show here that zebrafish gelsolin is required for proper dorsalization during embryogenesis. Inhibition of gelsolin expression by injecting fertilized eggs with a specific morpholino oligonucleotide resulted in a range of concentration-dependent ventralized phenotypes, including those lacking a brain and eyes. These were rescued by coinjection of zebrafish gelsolin or chordin (a known dorsalizing agent) mRNAs, or human gelsolin protein. Moreover, injection of gelsolin mRNA or human gelsolin protein by itself dorsalized the developing embryos, often resulting in axis duplication. Injection of the gelsolin-specific morpholino oligonucleotide enhanced the expression of Vent mRNA, a ventral marker downstream of bone morphogenetic proteins, whereas injection of gelsolin mRNA enhanced the expression of chordin and goosecoid mRNAs, both dorsal markers. Our results indicate that gelsolin also modulates embryonic dorsalventral pattern formation in zebrafish.


Subject(s)
Gelsolin/physiology , Intercellular Signaling Peptides and Proteins , Zebrafish/embryology , Zebrafish/physiology , Animals , Base Sequence , Body Patterning/genetics , Body Patterning/physiology , Bone Morphogenetic Proteins/metabolism , Cornea/embryology , Cornea/metabolism , Gelsolin/antagonists & inhibitors , Gelsolin/genetics , Gene Expression Regulation, Developmental , Glycoproteins/genetics , Homeodomain Proteins/genetics , In Situ Hybridization , Oligodeoxyribonucleotides, Antisense/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Signal Transduction , Zebrafish/genetics , Zebrafish Proteins/genetics
17.
Amyloid ; 9(2): 83-7, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12440480

ABSTRACT

Familial amyloidosis of the Finnish type (FAF) is caused by two proteolytic cleavages of mutant gelsolin leading to the accumulation of FAF amyloid in the patients' tissues. Here, we demonstrate that, in mouse pituitary corticotropic AtT20 cells, the enzyme responsible for the first cleavage of mutant secretory FAF gelsolin to FAF amyloid precursor is present in reasonable amounts. Furthermore, in At T20 cells stably expressing alpha1-PDX a potent inhibitor of most proprotein convertases, this cleavage was inhibited The present data provide strong evidence that proprotein convertases, possibly furin or PC5, are involved in the initialpathological cleavage of mutant secretory FAF gelsolin leading ultimately to the amyloid disease.


Subject(s)
Amyloid/metabolism , Amyloidosis, Familial/metabolism , Gelsolin/metabolism , Pituitary Gland/enzymology , Subtilisins/physiology , Animals , Cell Line , Furin , Gelsolin/antagonists & inhibitors , Humans , In Vitro Techniques , Mice , Pituitary Gland/cytology , Proprotein Convertases , Subtilisins/antagonists & inhibitors , Transfection , alpha 1-Antitrypsin/pharmacology
18.
Biochemistry ; 40(4): 904-13, 2001 Jan 30.
Article in English | MEDLINE | ID: mdl-11170411

ABSTRACT

Gelsolin, an actin and phosphoinositide binding protein, was photoaffinity labeled using a variety of benzophenone-containing phosphoinositide polyphosphate analogues. The N-terminal half and the C-terminal half of gelsolin showed synergy in the binding of phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P2]. Competitive displacement experiments with dibutyryl, dioctanoyl, or dipalmitoyl derivatives of PtdIns(4,5)P(2) suggested that, in addition to the inositol headgroup, a diacylglyceryl moiety was important for binding; these analogues also inhibited the gelsolin-severing activity of F-actin. In addition to the previously identified PtdIns(4,5)P2 binding site in the N-terminal half of gelsolin, a new binding site was identified in the C-terminal half by mapping the photocovalently modified peptide fragments. Moreover, increasing concentrations of Ca(2+) decreased the binding of the photolabile analogues to the C-terminal phosphoinositide binding site on gelsolin. A molecular model of the binding of PtdIns(4,5)P2 within two folded repeats of gelsolin has been calculated using these data.


Subject(s)
Gelsolin/metabolism , Peptide Fragments/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Photoaffinity Labels/metabolism , Actins/antagonists & inhibitors , Actins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Calcium/pharmacology , Dimerization , Gelsolin/antagonists & inhibitors , Gelsolin/chemistry , Horses , Humans , Models, Molecular , Molecular Sequence Data , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Mapping , Phosphatidylinositol 4,5-Diphosphate/analogs & derivatives , Phosphatidylinositol 4,5-Diphosphate/chemical synthesis , Phosphatidylinositol 4,5-Diphosphate/pharmacology , Phosphorylation , Polymers/metabolism , Protein Structure, Tertiary , Structure-Activity Relationship
19.
Mol Biol Cell ; 7(5): 743-53, 1996 May.
Article in English | MEDLINE | ID: mdl-8744948

ABSTRACT

Based on previous studies demonstrating activation of phosphatidylinositol 3-hydroxyl kinase (PI3-kinase) and stimulation of a change in cell shape, we examined the effect of osteopontin on the association of phospholipids with gelsolin, an actin-capping/severing protein. Osteopontin stimulated a rapid increase in phosphatidylinositol bisphosphate and phosphatidylinositol triphosphate levels associated with gelsolin in Triton-soluble fractions of cell lysates. The increased levels of phosphatidylinositol triphosphate associated with gelsolin were due to stimulation of PI3-kinase activity associated with gelsolin in the Triton-soluble fractions, and they were blocked by the PI3-kinase inhibitor wortmannin. Osteopontin stimulated translocation of PI3-kinase from the Triton-insoluble to Triton-soluble gelsolin. Osteopontin also decreased Triton-soluble gelsolin/actin complexes consistent with actin uncapping, and increased F-actin levels, which were also blocked by wortmannin. The osteopontin effects were mediated through binding to the alpha(v)beta 3 integrin. Taken together, our studies indicate that osteopontin/alpha(v)beta 3-mediated changes in gelsolin-associated phosphoinositide levels and PI3-kinase activity are related to stimulation of F-actin formation in osteoclasts.


Subject(s)
Gelsolin/metabolism , Osteoclasts/enzymology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Sialoglycoproteins/pharmacology , Actins/metabolism , Androstadienes/pharmacology , Animals , Blotting, Western , Chromatography, Thin Layer , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Female , Gelsolin/antagonists & inhibitors , Osteopontin , Phosphatidylinositol 3-Kinases , Phosphatidylinositol Phosphates/metabolism , Phospholipids/analysis , Phosphoproteins/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Precipitin Tests , Time Factors , Wortmannin
20.
J Biol Chem ; 269(52): 32916-23, 1994 Dec 30.
Article in English | MEDLINE | ID: mdl-7806519

ABSTRACT

We describe an assay for measuring both the extent and kinetics of the severing of F-actin, based on the enhanced fluorescence emission of tetramethylrhodamine-phalloidin bound to F-actin. The enhanced fluorescence is lost after exposure to active gelsolin by displacement of the phalloidin from actin during severing. This assay requires small amounts of actin and gelsolin, can be used to measure reaction times ranging from 1 to 10(3) s, and does not require covalent modification of either protein. The rate of fluorescence loss is linearly related to the concentrations of both actin and gelsolin. However, the apparent rate constant of the reaction is highly dependent on the divalent cation concentration, varying between 10(4) and 10(6) M-1 s-1 when the [Ca2+] varies between 20 and 200 microM. Addition of Mg2+ increases the apparent rate constant at equivalent Ca2+ concentration. These results suggest that in vitro the rate-limiting step in the severing process is the activation of gelsolin by the binding of Ca2+ and Mg2+ to several low affinity (Kd approximately 100 microM) sites on gelsolin. While activation of gelsolin by Ca2+ is a slow process, the binding and severing of actin occurs at a rate approaching the diffusion limit.


Subject(s)
Actins/metabolism , Calcium/metabolism , Gelsolin/physiology , Magnesium/metabolism , Phalloidine/metabolism , Animals , Coloring Agents , Gelsolin/antagonists & inhibitors , Kinetics , Rabbits , Rhodamines , Spectrometry, Fluorescence/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...