Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(24)2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34948078

ABSTRACT

Chronic kidney disease (CKD) is normally related to proteinuria, a common finding in a compromised glomerular filtration barrier (GFB). GFB is a structure composed of glomerular endothelial cells, the basement membrane, and the podocytes. CKD with podocyte damage may be associated with actin cytoskeleton reorganization, resulting in podocyte effacement. Gelsolin plays a critical role in several diseases, including cardiovascular diseases and cancer. Our current study aimed to determine the connection between gelsolin and podocyte, and thus the mechanism underlying podocyte injury in CKD. Experiments were carried out on Drosophila to demonstrate whether gelsolin had a physiological role in maintaining podocyte. Furthermore, the survival rate of gelsolin-knocked down Drosophila larvae was extensively reduced after AgNO3 exposure. Secondly, the in vitro podocytes treated with puromycin aminonucleoside (PAN) enhanced the gelsolin protein expression, as well as small GTPase RhoA and Rac1, which also regulated actin dynamic expression incrementally with the PAN concentrations. Thirdly, we further demonstrated in vivo that GSN was highly expressed inside the glomeruli with mitochondrial dysfunction in a CKD mouse model. Our findings suggest that an excess of gelsolin may contribute to podocytes damage in glomeruli.


Subject(s)
Gelsolin/physiology , Podocytes/metabolism , Renal Insufficiency, Chronic/metabolism , Animals , Animals, Genetically Modified , Disease Models, Animal , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Kidney Glomerulus/metabolism , Kidney Glomerulus/physiopathology , Mice , Podocytes/pathology , Renal Insufficiency, Chronic/physiopathology
2.
Sci Rep ; 11(1): 13197, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34162956

ABSTRACT

A combinatorial code of identity transcription factors (iTFs) specifies the diversity of muscle types in Drosophila. We previously showed that two iTFs, Lms and Ap, play critical role in the identity of a subset of larval body wall muscles, the lateral transverse (LT) muscles. Intriguingly, a small portion of ap and lms mutants displays an increased number of LT muscles, a phenotype that recalls pathological split muscle fibers in human. However, genes acting downstream of Ap and Lms to prevent these aberrant muscle feature are not known. Here, we applied a cell type specific translational profiling (TRAP) to identify gene expression signatures underlying identity of muscle subsets including the LT muscles. We found that Gelsolin (Gel) and dCryAB, both encoding actin-interacting proteins, displayed LT muscle prevailing expression positively regulated by, the LT iTFs. Loss of dCryAB function resulted in LTs with irregular shape and occasional branched ends also observed in ap and lms mutant contexts. In contrast, enlarged and then split LTs with a greater number of myonuclei formed in Gel mutants while Gel gain of function resulted in unfused myoblasts, collectively indicating that Gel regulates LTs size and prevents splitting by limiting myoblast fusion. Thus, dCryAB and Gel act downstream of Lms and Ap and contribute to preventing LT muscle branching and splitting. Our findings offer first clues to still unknown mechanisms of pathological muscle splitting commonly detected in human dystrophic muscles and causing muscle weakness.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/genetics , Gelsolin/physiology , Gene Expression Regulation , Genes, Insect , Muscles/ultrastructure , Muscular Dystrophy, Animal/genetics , alpha-Crystallin B Chain/physiology , Animals , Animals, Genetically Modified , CRISPR-Cas Systems , Cell Fusion , Cell Shape , Disease Models, Animal , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Embryo, Nonmammalian , Gelsolin/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Larva , Loss of Function Mutation , Multigene Family , Muscle Cells/metabolism , Muscles/metabolism , Muscular Dystrophy, Animal/pathology , Myoblasts/metabolism , Myoblasts/ultrastructure , RNA, Messenger/metabolism , Transcription Factors/physiology , Transcription, Genetic , alpha-Crystallin B Chain/genetics
3.
Dev Biol ; 469: 12-25, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32980309

ABSTRACT

The sarcomere is the basic contractile unit of muscle, composed of repeated sets of actin thin filaments and myosin thick filaments. During muscle development, sarcomeres grow in size to accommodate the growth and function of muscle fibers. Failure in regulating sarcomere size results in muscle dysfunction; yet, it is unclear how the size and uniformity of sarcomeres are controlled. Here we show that the formin Diaphanous is critical for the growth and maintenance of sarcomere size: Dia sets sarcomere length and width through regulation of the number and length of the actin thin filaments in the Drosophila flight muscle. To regulate thin filament length and sarcomere size, Dia interacts with the Gelsolin superfamily member Flightless I (FliI). We suggest that these actin regulators, by controlling actin dynamics and turnover, generate uniformly sized sarcomeres tuned for the muscle contractions required for flight.


Subject(s)
Drosophila Proteins/physiology , Formins/physiology , Gelsolin/physiology , Sarcomeres/ultrastructure , Animals , Drosophila/genetics , Drosophila/physiology , Drosophila/ultrastructure , Drosophila Proteins/genetics , Flight, Animal , Formins/genetics , Gene Knockdown Techniques , Muscles/ultrastructure
4.
Int J Mol Sci ; 21(19)2020 Oct 06.
Article in English | MEDLINE | ID: mdl-33036217

ABSTRACT

The emergent properties of the array arrangement of the molecular motor myosin II in the sarcomere of the striated muscle, the generation of steady force and shortening, can be studied in vitro with a synthetic nanomachine made of an ensemble of eight heavy-meromyosin (HMM) fragments of myosin from rabbit psoas muscle, carried on a piezoelectric nanopositioner and brought to interact with a properly oriented actin filament attached via gelsolin (a Ca2+-regulated actin binding protein) to a bead trapped by dual laser optical tweezers. However, the application of the original version of the nanomachine to investigate the Ca2+-dependent regulation mechanisms of the other sarcomeric (regulatory or cytoskeleton) proteins, adding them one at a time, was prevented by the impossibility to preserve [Ca2+] as a free parameter. Here, the nanomachine is implemented by assembling the bead-attached actin filament with the Ca2+-insensitive gelsolin fragment TL40. The performance of the nanomachine is determined both in the absence and in the presence of Ca2+ (0.1 mM, the concentration required for actin attachment to the bead with gelsolin). The nanomachine exhibits a maximum power output of 5.4 aW, independently of [Ca2+], opening the possibility for future studies of the Ca2+-dependent function/dysfunction of regulatory and cytoskeletal proteins.


Subject(s)
Calcium/metabolism , Muscle Contraction , Myosin Type II/metabolism , Nanostructures/chemistry , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/physiology , Animals , Gelsolin/metabolism , Gelsolin/physiology , Male , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Myosin Type II/physiology , Rabbits
5.
Br J Haematol ; 190(3): 418-429, 2020 08.
Article in English | MEDLINE | ID: mdl-32130729

ABSTRACT

Myelodysplastic syndrome (MDS), a largely incurable hematological malignancy, is driven by complex genetic and epigenetic alterations from an aberrant clone of hematopoietic stem/progenitor cells (HSPCs). Ubiquitin-specific protease 7 (USP7) has been demonstrated to have an important oncogenic role in the development of several cancer types, but its role in MDS is unknown. Here, we demonstrate that USP7 expression is elevated in MDS cell lines and patient samples. The USP7-selective small-molecule inhibitors P5091 and P22077 inhibited cell proliferation and induced megakaryocytic differentiation in both cell lines and primary cells. Furthermore, pharmacological inhibition of USP7 markedly suppressed the growth of MDS cell lines in xenograft mouse models. To explore the mechanisms underlying the observed phenotypic changes, we employed RNA-seq to compare the differences in genes after USP7 inhibitor treatment and found that gelsolin (GSN) expression was increased significantly after USP7 inhibitor treatment. Furthermore, knockdown of GSN attenuated the proliferation inhibition, apoptosis induction and megakaryocyte differentiation induced by USP7 inhibitors in MDS cells. Collectively, our findings identify previously unknown roles of USP7 and suggest that the USP7/GSN axis may be a potential therapeutic target in MDS.


Subject(s)
Gelsolin/physiology , Megakaryocytes/drug effects , Myelodysplastic Syndromes/pathology , Protease Inhibitors/pharmacology , Thiophenes/pharmacology , Thrombopoiesis/drug effects , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line/transplantation , Enzyme Induction/drug effects , Gelsolin/biosynthesis , Gelsolin/genetics , Heterografts , Humans , Megakaryocytes/pathology , Mice , Mice, Inbred NOD , Neoplasms, Experimental/etiology , Risk , Transcriptome/drug effects , Ubiquitin-Specific Peptidase 7/physiology , Up-Regulation/drug effects
6.
Nat Methods ; 16(11): 1095-1100, 2019 11.
Article in English | MEDLINE | ID: mdl-31611691

ABSTRACT

Intracellular antibodies have become powerful tools for imaging, modulating and neutralizing endogenous target proteins. Here, we describe an optogenetically activated intracellular antibody (optobody) consisting of split antibody fragments and blue-light inducible heterodimerization domains. We expanded this optobody platform by generating several optobodies from previously developed intracellular antibodies, and demonstrated that photoactivation of gelsolin and ß2-adrenergic receptor (ß2AR) optobodies suppressed endogenous gelsolin activity and ß2AR signaling, respectively.


Subject(s)
Antibodies/physiology , Gelsolin/physiology , Optogenetics , Receptors, Adrenergic, beta-2/physiology , Animals , Cells, Cultured , Humans
7.
Sci Rep ; 8(1): 12602, 2018 08 22.
Article in English | MEDLINE | ID: mdl-30135452

ABSTRACT

Here, we report that minimal functional gelsolin i.e. fragment 28-161 can display F-actin depolymerizing property even after heating the protein to 80 °C. Small angle X-ray scattering (SAXS) data analysis confirmed that under Ca2+-free conditions, 28-161 associates into monomer to dimer and tetramer, which later forms ß-amyloids, but in presence of Ca2+, it forms dimers which proceed to non-characterizable aggregates. The dimeric association also explained the observed decrease in ellipticity in circular dichroism experiments with increase in temperature. Importantly, SAXS data based models correlated well with our crystal structure of dimeric state of 28-161. Characterization of higher order association by electron microscopy, Congo red and ThioflavinT staining assays further confirmed that only in absence of Ca2+ ions, heating transforms 28-161 into ß-amyloids. Gel filtration and other experiments showed that ß-amyloids keep leaching out the monomer, and the release rates could be enhanced by addition of L-Arg to the amyloids. F-actin depolymerization showed that addition of Ca2+ ions to released monomer initiated the depolymerization activity. Overall, we propose a way to compose a supramolecular assembly which releases functional protein in sustained manner which can be applied for varied potentially therapeutic interventions.


Subject(s)
Actins/metabolism , Gelsolin/metabolism , Actin Cytoskeleton , Actin Depolymerizing Factors/metabolism , Amyloid beta-Peptides/metabolism , Crystallography, X-Ray , Gelsolin/physiology , Hot Temperature , Models, Molecular , Protein Binding , Protein Denaturation , Temperature , X-Ray Diffraction
8.
Technol Cancer Res Treat ; 16(4): 512-518, 2017 08.
Article in English | MEDLINE | ID: mdl-27121073

ABSTRACT

Gelsolin is an actin-binding protein and acts as an important regulator of cell survival. This study aimed to determine the function of gelsolin in the radioresistance of non-small cell lung cancer cells. We examined the expression of gelsolin in radioresistant A549 and H460 cells and their parental cells. The effects of gelsolin overexpression and knockdown on the clonogenic survival and apoptosis of non-small cell lung cancer cells after irradiation were studied. The involvement of phosphoinositide 3-kinase/Akt signaling in the action of gelsolin was checked. We found that gelsolin was significantly upregulated in radioresistant A549 and H460 cells. Overexpression of gelsolin significantly ( P < .05) increased the number of colonies from irradiated A549 and H460 cells compared to transfection of empty vector. In contrast, knockdown of gelsolin significantly ( P < .05) suppressed colony formation after irradiation. Gelsolin-overexpressing cells displayed reduced apoptosis in response to irradiation, which was coupled with decreased levels of cleaved caspase-3 and poly adenosine diphosphate-ribose polymerase. Ectopic expression of gelsolin significantly ( P < .05) enhanced the phosphorylation of Akt compared to nontransfected cells. Pretreatment with the phosphoinositide 3-kinase inhibitor LY294002 (20 µmol/L) significantly decreased clonogenic survival and enhanced apoptosis in gelsolin-overexpressing A549 and H460 cells after irradiation. Taken together, gelsolin upregulation promotes radioresistance in non-small cell lung cancer cells, at least partially, through activation of phosphoinositide 3-kinase/Akt signaling.


Subject(s)
Carcinoma, Non-Small-Cell Lung/radiotherapy , Gelsolin/physiology , Lung Neoplasms/radiotherapy , Radiation Tolerance , Signal Transduction , A549 Cells , Apoptosis , Carcinoma, Non-Small-Cell Lung/metabolism , Gene Expression , Humans , Lung Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Up-Regulation
9.
Gene ; 571(2): 292-7, 2015 Oct 25.
Article in English | MEDLINE | ID: mdl-26149653

ABSTRACT

OBJECTIVE: Gelsolin (GSN), one of the most important actin structure regulating proteins, has been implicated in the oncogenesis of some cancers. In this study, we investigated the expression of GSN in hepatocellular carcinoma (HCC) and revealed its potential mechanisms. The mRNA and protein levels of GSN were overexpressed in HCC cells and HCC tissues compared to adjacent noncancerous tissues. GSN expression was correlated with venous invasion (P=0.0199) and Edmonson grading (P=0.0344) expression in HCC. Overexpression of GSN in Huh7 and SMMC-7721 cells significantly promoted cell proliferation and the number of Matrigel™-invading cells compared with control cells, with increased expression of matrix metalloproteinase MCL-1, MMP-2 and MMP-9, a key regulator of growth and invasion. In contrast, knockdown of GSN expression with small interfering RNA (siRNA) in MHCC-97L and MHCC-97H cell lines resulted in decreased cell viability and cell invasion. Our findings indicated that GSN expression promoted tumor-associated phenotypes by facilitating proliferative and invasive capacities of HCC cells, which might serve as a potential therapeutic target for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Proliferation/genetics , Gelsolin/physiology , Liver Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Female , Gelsolin/genetics , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Middle Aged , Neoplasm Invasiveness , Tumor Cells, Cultured
10.
J Physiol Pharmacol ; 66(3): 373-83, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26084219

ABSTRACT

Ion channels involved in cardiac excitation-contraction coupling are linked to the cytoskeleton. Therefore changes in the cytoskeletal actin filaments may influence cardiac membrane currents and electro-mechanical coupling. Depolymerization of actin filaments by gelsolin (gsn) is involved in the organisation of the cytoskeleton by leading to a lower polymerization state. Gsn is activated by Ca(2+) and inhibited by phosphoinositol-bisphosphate (PIP2). Furthermore, gsn has been linked to pathological conditions with reduced contractility like heart failure, amyloidosis and apoptosis. Thus, we hypothesize, that gsn deficiency may change electromechanical properties of freshly isolated ventricular cardiomyocytes. We recorded L-type Ca(2+) current (ICa,L) in whole-cell patch clamp mode in freshly isolated ventricular cardiomyocytes from gsn deficient ((-/-)) and control (gsn(+/+)) mice. Sarcomere shortening was monitored in field-stimulated myocytes from 0.5 Hz to 10 Hz by video microscopy. Shortening-frequency relation, post-rest potentiation and ß-adrenergic stimulation were investigated. ICa,L was increased in gsn(-/-) vs. gsn(+/+) myocytes. Sarcomere shortening amplitude and velocity were enhanced in gsn(-/-) vs. gsn(+/+) at all frequencies. Shortening-frequency relationship showed a biphasic pattern with decay in shortening amplitude between 0.5 and 2 Hz and an increase at higher frequencies in both genotypes. Post-rest characteristics revealed a frequency-dependent decay of post-rest potentiation in gsn(+/+) while it remained stable in gsn(-/-). In gsn(-/-) a reduced response to ß-adrenergic stimulation was observed. Resting sarcomere length was shorter in gsn(-/-) but neither increasing frequency nor ß-adrenergic stimulation induced further decay in any of the genotypes. In summary, gsn deficiency had a profound effect on excitiation-contraction properties and improved systolic function while not affecting diastolic function in unloaded isolated cardiomyocytes. Therefore, gsn mediated effects on contractility may play a role in patients with heart failure and cancer, where gsn levels are known to be elevated.


Subject(s)
Gelsolin/physiology , Myocytes, Cardiac/physiology , Animals , Calcium Channels, L-Type/physiology , Excitation Contraction Coupling , Female , Gelsolin/deficiency , Gelsolin/genetics , Heart/anatomy & histology , Male , Mice, Knockout , Sarcomeres/physiology
11.
FASEB J ; 29(6): 2281-91, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25681458

ABSTRACT

Osteoclast differentiation and function are highly dependent on the assembly and turnover of actin filaments, but little is known about the roles of actin binding proteins in these processes. Adseverin (Ads), a member of the gelsolin superfamily of actin capping and severing proteins, regulates actin filament turnover and can regulate the turnover of cortical actin filaments of chromaffin cells during exocytosis. Using a conditional Ads knockout mouse model, we confirmed our previous finding in cultured cells that Ads plays a role in osteoclastogenesis (OCG) and actin cytoskeletal organization in osteoclasts. Here we show that Ads is required for osteoclast formation and that when alveolar bone resorption is experimentally induced in mice, genetic deletion of Ads prevents osteoclast-mediated bone loss. Further, when Ads-null osteoclasts are cultured, they exhibit defective OCG, disorganized podosome-based actin filament superstructures, and decreased bone resorption. Reintroduction of Ads into Ads-null osteoclast precursor cells restored these osteoclast defects. Collectively, these data demonstrate a unique and osteoclast-specific role for Ads in OCG and osteoclast function.


Subject(s)
Alveolar Bone Loss/physiopathology , Cell Differentiation/physiology , Gelsolin/physiology , Osteoclasts/metabolism , Periodontal Diseases/physiopathology , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Alveolar Bone Loss/genetics , Animals , Blotting, Western , Cell Differentiation/genetics , Cells, Cultured , Fluorescence Recovery After Photobleaching , Gelsolin/deficiency , Gelsolin/genetics , Mice, Knockout , Microscopy, Confocal , Osteoclasts/cytology , Periodontal Diseases/genetics , Transfection
12.
J Endod ; 41(4): 493-500, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25583547

ABSTRACT

INTRODUCTION: Adseverin is an actin-severing and actin-capping protein that is primarily expressed in secretory cells, where it regulates the filamentous actin cytoskeleton during cell differentiation and exocytosis. However, little is known regarding its regulatory role in dental pulp cells (DPCs). This study examined the expression and function of adseverin in the proliferation, migration, and odontoblastic differentiation of DPCs. METHODS: DPCs were assayed for morphologic changes, proliferation, migration, alkaline phosphatase activity, and dentin sialoprotein and dentin matrix protein-1 protein levels in vitro after knockdown of adseverin by using small interfering RNA. Tooth germs isolated from Sprague-Dawley rats were processed for immunohistochemistry analysis of adseverin. RESULTS: Adseverin expression was increased in a time-dependent fashion in the early stage of odontoblastic differentiation. When adseverin expression was suppressed in DPCs, their cellular morphology was altered, and their proliferation, migration, and odontoblastic differentiation were substantially decreased in vitro. Secretory odontoblasts in the tooth germ at day 5 post partum expressed a stronger adseverin signal compared with those at days 1 and 3 post partum. CONCLUSIONS: Adseverin may play a crucial role in the proliferation, migration, and odontoblastic differentiation of DPCs via filamentous actin cytoskeleton regulation. However, further investigations are required to clarify the underlying mechanisms.


Subject(s)
Dental Pulp/cytology , Gelsolin/physiology , Odontoblasts/cytology , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , Gelsolin/biosynthesis , Humans
13.
Asian J Androl ; 14(6): 816-21, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23001443

ABSTRACT

Mammalian sperm must undergo a series of biochemical and physiological modifications, collectively called capacitation, in the female reproductive tract prior to the acrosome reaction (AR). The mechanisms of these modifications are not well characterized though protein kinases were shown to be involved in the regulation of intracellular Ca(2+) during both capacitation and the AR. In the present review, we summarize some of the signaling events that are involved in capacitation. During the capacitation process, phosphatidyl-inositol-3-kinase (PI3K) is phosphorylated/activated via a protein kinase A (PKA)-dependent cascade, and downregulated by protein kinase C α (PKCα). PKCα is active at the beginning of capacitation, resulting in PI3K inactivation. During capacitation, PKCα as well as PP1γ2 is degraded by a PKA-dependent mechanism, allowing the activation of PI3K. The activation of PKA during capacitation depends mainly on cyclic adenosine monophosphate (cAMP) produced by the bicarbonate-dependent soluble adenylyl cyclase. This activation of PKA leads to an increase in actin polymerization, an essential process for the development of hyperactivated motility, which is necessary for successful fertilization. Actin polymerization is mediated by PIP(2) in two ways: first, PIP(2) acts as a cofactor for phospholipase D (PLD) activation, and second, as a molecule that binds and inhibits actin-severing proteins such as gelsolin. Tyrosine phosphorylation of gelsolin during capacitation by Src family kinase (SFK) is also important for its inactivation. Prior to the AR, gelsolin is released from PIP(2) and undergoes dephosphorylation/activation, resulting in fast F-actin depolymerization, leading to the AR.


Subject(s)
Acrosome Reaction/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , Sperm Capacitation/physiology , src-Family Kinases/metabolism , Actins/metabolism , Animals , Bicarbonates/metabolism , Calcium/metabolism , Female , Gelsolin/physiology , Humans , Male , Phosphorylation , Protein Kinase C-alpha/metabolism , Protein Processing, Post-Translational , Tyrosine/metabolism
14.
J Immunol ; 189(2): 629-37, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22706076

ABSTRACT

The neutrophil formyl peptide receptors, FPR1 and FPR2, play critical roles for inflammatory reactions, and receptor-specific antagonists/inhibitors can possibly be used to facilitate the resolution of pathological inflammatory reactions. A 10-aa-long rhodamine-linked and membrane-permeable peptide inhibitor (PBP10) has such a potential. This FPR2 selective inhibitor adopts a phosphatidylinositol 4,5-bisphosphate-binding sequence in the cytoskeletal protein gelsolin. A core peptide, RhB-QRLFQV, is identified that displays inhibitory effects as potent as the full-length molecule. The phosphatidylinositol 4,5-bisphosphate-binding capacity of PBP10 was not in its own sufficient for inhibition. A receptor in which the presumed cytoplasmic signaling C-terminal tail of FPR2 was replaced with that of FPR1 retained the PBP10 sensitivity, suggesting that the tail of FPR2 was not on its own critical for inhibition. This gains support from the fact that the effect of cell-penetrating lipopeptide (a pepducin), suggested to act primarily through the third intracellular loop of FPR2, was significantly inhibited by PBP10. The third intracellular loops of FPR1 and FPR2 differ in only two amino acids, but an FPR2 mutant in which these two amino acids were replaced by those present in FPR1 retained the PBP10 sensitivity. In summary, we conclude that the inhibitory activity on neutrophil function of PBP10 is preserved in the core sequence RhB-QRLFQV and that neither the third intracellular loop of FPR2 nor the cytoplasmic tail of the receptor alone is responsible for the specific inhibition.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Gelsolin/chemistry , Gelsolin/physiology , Peptides/chemistry , Peptides/physiology , Receptors, Formyl Peptide/chemistry , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/chemistry , Receptors, Lipoxin/physiology , Amino Acid Sequence , Cell Membrane Permeability/immunology , Dose-Response Relationship, Immunologic , Gelsolin/metabolism , HL-60 Cells , Humans , Molecular Sequence Data , Neutrophil Activation/immunology , Peptides/metabolism , Protein Binding/immunology , Protein Structure, Tertiary , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism
15.
J Immunol ; 188(7): 3237-46, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22387556

ABSTRACT

B cells encounter both soluble Ag (sAg) and membrane-associated Ag (mAg) in the secondary lymphoid tissue, yet how the physical form of Ag modulates B cell activation remains unclear. This study compares actin reorganization and its role in BCR signalosome formation in mAg- and sAg-stimulated B cells. Both mAg and sAg induce F-actin accumulation and actin polymerization at BCR microclusters and at the outer rim of BCR central clusters, but the kinetics and magnitude of F-actin accumulation in mAg-stimulated B cells are greater than those in sAg-stimulated B cells. Accordingly, the actin regulatory factors, cofilin and gelsolin, are recruited to BCR clusters in both mAg- and sAg-stimulated B cells but with different kinetics and patterns of cellular redistribution. Inhibition of actin reorganization by stabilizing F-actin inhibits BCR clustering and tyrosine phosphorylation induced by both forms of Ag. Depolymerization of F-actin leads to unpolarized microclustering of BCRs and tyrosine phosphorylation in BCR microclusters without mAg and sAg, but with much slower kinetics than those induced by Ag. Therefore, actin reorganization, mediated via both polymerization and depolymerization, is required for the formation of BCR signalosomes in response to both mAg and sAg.


Subject(s)
Actin Cytoskeleton/ultrastructure , Actins/chemistry , Antigens/immunology , Cytoskeleton/ultrastructure , Lymphocyte Activation/immunology , Receptors, Antigen, B-Cell/immunology , Actin Depolymerizing Factors/physiology , Animals , Biopolymers , Cell Membrane/immunology , Cell Polarity , Gelsolin/physiology , Mice , Mice, Inbred CBA , Phosphorylation , Protein Processing, Post-Translational , Protein Structure, Quaternary , Solubility
16.
PLoS One ; 7(1): e29408, 2012.
Article in English | MEDLINE | ID: mdl-22238609

ABSTRACT

Plasmodium sporozoites are transmitted by Anopheles mosquitoes and infect hepatocytes, where a single sporozoite replicates into thousands of merozoites inside a parasitophorous vacuole. The nature of the Plasmodium-host cell interface, as well as the interactions occurring between these two organisms, remains largely unknown. Here we show that highly dynamic hepatocyte actin reorganization events occur around developing Plasmodium berghei parasites inside human hepatoma cells. Actin reorganization is most prominent between 10 to 16 hours post infection and depends on the actin severing and capping protein, gelsolin. Live cell imaging studies also suggest that the hepatocyte cytoskeleton may contribute to parasite elimination during Plasmodium development in the liver.


Subject(s)
Actins/metabolism , Hepatocytes/parasitology , Plasmodium/metabolism , Protein Multimerization/physiology , Actin Cytoskeleton/metabolism , Animals , Cells, Cultured , Gelsolin/metabolism , Gelsolin/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hepatocytes/metabolism , Host-Parasite Interactions/genetics , Host-Parasite Interactions/physiology , Humans , Kinetics , Liver/metabolism , Liver/parasitology , Mice , Mice, Inbred BALB C , Organisms, Genetically Modified , Plasmodium/genetics , Plasmodium/physiology , Tubulin/metabolism
17.
BJOG ; 118(2): 145-53, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21054766

ABSTRACT

BACKGROUND: Immune responses to infection are uniquely regulated during gestation to allow for antimicrobial defence and tissue repair, whilst preventing damage to developing fetal organs or the triggering of preterm labour. OBJECTIVE: A review and analysis of studies delineating gestation-specific immune modulation and intra-amniotic regulation of pro-inflammatory immunity. SEARCH STRATEGY: Identification of the alterations between the fetus/neonate and adult with regard to the endogenous and infection-induced expression of molecules with immune regulatory properties, and the characterisation of intra-amniotic immune mediators that inhibit bacterial-induced pro-inflammatory cytokine production. SELECTION CRITERIA: English and non-English publications from 1985 to the present. DATA COLLECTION AND ANALYSIS: An electronic literature search using MEDLINE, PubMed, articles cited in the primary sources, as well as pregnancy-related immunology research from our laboratory at Weill Medical College of Cornell University. MAIN RESULTS: During fetal development, interleukin (IL)-23, IL-10 and IL-6, as well as T-helper-17 (Th17)-mediated immune responses, are upregulated, whereas tumour necrosis factor-α (TNF-α) and IL-1ß- and Th1-mediated immune responses are downregulated in the intrauterine environment (both the fetal compartment and the amniotic compartment). Infection-related immunity during gestation is preferentially directed towards combating extracellular microbial pathogens. Amniotic fluid and the neonatal circulation contain multiple components that improve the ability of the developing neonate to tolerate microbial-induced immune activation. CONCLUSIONS: The repertoire of immune mechanisms to control infection and inflammation differ between fetal and adult life. The dual mechanisms of resistance to infection and tolerance to infection-induced immune activation prevent damage to the developing fetus and the triggering of premature labour.


Subject(s)
Cytokines/physiology , Fetus/immunology , Immunity, Cellular/physiology , Obstetric Labor, Premature/immunology , Pregnancy Complications, Infectious/immunology , Adenosine/physiology , Adult , Cytokines/biosynthesis , Cytokines/immunology , Exosomes/physiology , Female , Gelsolin/physiology , Histones/physiology , Humans , Hyaluronic Acid/physiology , Immunologic Factors/physiology , Neutrophils/physiology , Obstetric Labor, Premature/microbiology , Pregnancy , Up-Regulation
18.
Postepy Hig Med Dosw (Online) ; 64: 303-9, 2010 Jun 18.
Article in Polish | MEDLINE | ID: mdl-20558869

ABSTRACT

Gelsolin is an actin-binding and an actin-fragmenting protein. It contains 730 amino-acids, organized in six G1-G6 homologous domains which determine different functions of the protein. Two variants of gelsolin, cytoplasmic and secreted (contained in plasma) are described. Cytoplasmic gelsolin re-organizes the structure of cytoskeleton and plays an important role as a capping protein. In addition, cytoplasmic gelsolin binds bacterial lipopolysaccharide and ATP and exhibits antibacterial and anti-inflammatory properties. Plasma gelsolin is synthesized mainly in skeletal and smooth muscles and myocardium. Plasma gelsolin was also found in: blood, lymph, bronchial epithelia, synovial fluids and cerebro-spinal fluid. The protein plays a role in the immune response, moreover it is involved in extracellular and blood actin-scavenger system. Plasma gelsolin has anti-amyloidogenic, anti-oxidant and anti-apoptotic properties and it has a potential for treatment of Alzheimer disease. Decreased levels of the gelsolin plasma isoform was observed in patients with sepsis, myocardial infarction, liver failure, acute respiratory distress syndrome, inflammations and after burns. On the other hand, after rhabdomyolysis and in amyloidosis gelsolin plasma level are increased. In this review we present recent data on the structure and functions of gelsolin and changes of its activity in some pathological processes.


Subject(s)
Gelsolin/chemistry , Gelsolin/physiology , Humans
19.
Biochemistry ; 49(20): 4349-60, 2010 May 25.
Article in English | MEDLINE | ID: mdl-20392036

ABSTRACT

Caenorhabditis elegans gelsolin-like protein-1 (GSNL-1) is a new member of the gelsolin family of actin regulatory proteins [Klaavuniemi, T., Yamashiro, S., and Ono, S. (2008) J. Biol. Chem. 283, 26071-26080]. It is an unconventional gelsolin-related protein with four gelsolin-like (G) domains (G1-G4), unlike typical gelsolin-related proteins with three or six G domains. GSNL-1 severs actin filaments and caps the barbed end in a calcium-dependent manner similar to that of gelsolin. In contrast, GSNL-1 has properties different from those of gelsolin in that it remains bound to F-actin and does not nucleate actin polymerization. To understand the mechanism by which GSNL-1 regulates actin dynamics, we investigated the domain-function relationship of GSNL-1 by analyzing activities of truncated forms of GSNL-1. G1 and the linker between G1 and G2 were sufficient for actin filament severing, whereas G1 and G2 were required for barbed end capping. The actin severing activity of GSNL-1 was inhibited by phosphatidylinositol 4,5-bisphosphate (PIP2), and a PIP2-sensitive domain was mapped to G1 and G2. At least two actin-binding sites were detected: a calcium-dependent G-actin-binding site in G1 and a calcium-independent G- and F-actin-binding site in G3 and G4. These results reveal both conserved and different utilization of G domains between C. elegans GSNL-1 and mammalian gelsolin for actin regulatory functions.


Subject(s)
Actin Capping Proteins/metabolism , Actin Cytoskeleton/metabolism , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/metabolism , Intracellular Calcium-Sensing Proteins/chemistry , Intracellular Calcium-Sensing Proteins/metabolism , Phosphatidylinositols/metabolism , Actin Capping Proteins/chemistry , Actin Capping Proteins/physiology , Actin Depolymerizing Factors/chemistry , Actin Depolymerizing Factors/genetics , Actin Depolymerizing Factors/metabolism , Actin Depolymerizing Factors/physiology , Actins/metabolism , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/physiology , Gelsolin/chemistry , Gelsolin/metabolism , Gelsolin/physiology , Intracellular Calcium-Sensing Proteins/genetics , Intracellular Calcium-Sensing Proteins/physiology , Models, Biological , Molecular Weight , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Binding/physiology , Protein Interaction Mapping , Protein Structure, Tertiary/physiology
20.
Proteomics ; 10(6): 1316-27, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20127684

ABSTRACT

Global protein expression profiling can potentially uncover perturbations associated with common forms of heart disease. We have used shotgun MS/MS to monitor the state of biological systems in cardiac tissue correlating with disease onset, cardiac insufficiency and progression to heart failure in a time-course mouse model of dilated cardiomyopathy. However, interpreting the functional significance of the hundreds of differentially expressed proteins has been challenging. Here, we utilize improved enrichment statistical methods and an extensive collection of functionally related gene sets, gaining a more comprehensive understanding of the progressive alterations associated with functional decline in dilated cardiomyopathy. We visualize the enrichment results as an Enrichment Map, where significant gene sets are grouped based on annotation similarity. This approach vastly simplifies the interpretation of the large number of enriched gene sets found. For pathways of specific interest, such as Apoptosis and the MAPK (mitogen-activated protein kinase) cascade, we performed a more detailed analysis of the underlying signaling network, including experimental validation of expression patterns.


Subject(s)
Apoptosis/physiology , Cardiomyopathy, Dilated/physiopathology , Gene Expression Profiling , MAP Kinase Signaling System/physiology , Animals , Cardiomyopathy, Dilated/drug therapy , Cardiomyopathy, Dilated/genetics , Caspase 3/physiology , Databases, Protein , Gelsolin/physiology , Metabolomics , Mice , Propranolol/therapeutic use , Proteomics/methods , Systems Biology , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...