Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
1.
Autophagy ; 19(6): 1678-1692, 2023 06.
Article in English | MEDLINE | ID: mdl-36409297

ABSTRACT

Mitophagy is a form of autophagy that selectively removes damaged mitochondria and attenuates mitochondrial-dependent apoptosis during viral infection, but how arboviruses balance mitophagy and apoptosis to facilitate persistent viral infection in insect vectors without causing evident fitness cost remains elusive. Here, we identified mitochondrial VDAC1 (voltage-dependent anion channel 1) that could be hijacked by nonstructural protein Pns11 of rice gall dwarf virus (RGDV), a plant nonenveloped double-stranded RNA virus, to synergistically activate pro-viral extensive mitophagy and limited apoptosis in leafhopper vectors. The direct target of fibrillar structures constructed by Pns11 with VDAC1 induced mitochondrial degeneration. Moreover, the degenerated mitochondria were recruited into Pns11-induced phagophores to initiate mitophagy via interaction of VDAC1 with Pns11 and an autophagy protein, ATG8. Such mitophagy mediated by Pns11 and VDAC1 required the classical PRKN/Parkin-PINK1 pathway. VDAC1 regulates apoptosis by controlling the release of apoptotic signaling molecules through its pore, while the anti-apoptotic protein GSN (gelsolin) could bind to VDAC1 pore. We demonstrated that the interaction of Pns11 with VDAC1 and gelsolin decreased VDAC1 expression but increased GSN expression, which prevented the extensive apoptotic response in virus-infected regions. Meanwhile, virus-induced mitophagy also effectively prevented extensive apoptotic response to decrease apoptosis-caused insect fitness cost. The subsequent fusion of virus-loaded mitophagosomes with lysosomes is prevented, and thus such mitophagosomes are exploited for persistent spread of virions within insect bodies. Our results reveal a new strategy for arboviruses to balance and exploit mitophagy and apoptosis, resulting in an optimal intracellular environment for persistent viral propagation in insect vectors.Abbreviations: ATG: autophagy related; BNIP3: BCL2 interacting protein 3; CYCS/CytC: cytochrome c, somatic; dsGSN: double-stranded RNAs targeting GSN/gelsolin; dsGFP: double-stranded RNAs targeting green fluorescent protein; dsPRKN: double-stranded RNAs targeting PRKN; dsPns11: double-stranded RNAs targeting Pns11; dsRNA: double-stranded RNA; EC: epithelia cell; GST: glutathione S-transferase; LAMP1: lysosomal associated membrane protein 1; Mito: mitochondrion; Mmg: middle midgut; MP, mitophagosome; PG, phagophore. padp: post-first access to diseased plants; PINK1: PTEN induced kinase 1; RGDV: rice gall dwarf virus; SQSTM1: sequestosome 1; TOMM20: translocase of outer mitochondrial membrane 20; TUNEL: terminal deoxynucleotidyl transferase dUTP nick end labeling; VDAC1: voltage dependent anion channel 1.


Subject(s)
Arbovirus Infections , Hemiptera , Animals , Mitophagy/genetics , Hemiptera/genetics , Hemiptera/metabolism , Voltage-Dependent Anion Channel 1/genetics , RNA, Double-Stranded/pharmacology , Gelsolin/genetics , Gelsolin/metabolism , Gelsolin/pharmacology , Autophagy , Apoptosis , Protein Kinases/metabolism
2.
J Neuroinflammation ; 19(1): 282, 2022 Nov 24.
Article in English | MEDLINE | ID: mdl-36434734

ABSTRACT

BACKGROUND: Plasma gelsolin (pGSN) is an important part of the blood actin buffer that prevents negative consequences of possible F-actin deposition in the microcirculation and has various functions during host immune response. Recent reports reveal that severe COVID-19 correlates with reduced levels of pGSN. Therefore, using an in vitro system, we investigated whether pGSN could attenuate increased permeability of the blood-brain barrier (BBB) during its exposure to the portion of the SARS-CoV-2 spike protein containing the receptor binding domain (S1 subunit). MATERIALS AND METHODS: Two- and three-dimensional models of the human BBB were constructed using the human cerebral microvascular endothelial cell line hCMEC/D3 and exposed to physiologically relevant shear stress to mimic perfusion in the central nervous system (CNS). Trans-endothelial electrical resistance (TEER) as well as immunostaining and Western blotting of tight junction (TJ) proteins assessed barrier integrity in the presence of the SARS-CoV-2 spike protein and pGSN. The IncuCyte Live Imaging system evaluated the motility of the endothelial cells. Magnetic bead-based ELISA was used to determine cytokine secretion. Additionally, quantitative real-time PCR (qRT-PCR) revealed gene expression of proteins from signaling pathways that are associated with the immune response. RESULTS: pGSN reversed S1-induced BBB permeability in both 2D and 3D BBB models in the presence of shear stress. BBB models exposed to pGSN also exhibited attenuated pro-inflammatory signaling pathways (PI3K, AKT, MAPK, NF-κB), reduced cytokine secretion (IL-6, IL-8, TNF-α), and increased expression of proteins that form intercellular TJ (ZO-1, occludin, claudin-5). CONCLUSION: Due to its anti-inflammatory and protective effects on the brain endothelium, pGSN has the potential to be an alternative therapeutic target for patients with severe SARS-CoV-2 infection, especially those suffering neurological complications of COVID-19.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Spike Glycoprotein, Coronavirus , Blood-Brain Barrier , Gelsolin/pharmacology , Endothelial Cells , Permeability , Tight Junction Proteins , Cytokines
3.
Int J Mol Sci ; 21(7)2020 Apr 07.
Article in English | MEDLINE | ID: mdl-32272559

ABSTRACT

Plasma gelsolin (pGSN) is a highly conserved abundant circulating protein, characterized by diverse immunomodulatory activities including macrophage activation and the ability to neutralize pro-inflammatory molecules produced by the host and pathogen. Using a murine model of Gram-negative sepsis initiated by the peritoneal instillation of Pseudomonas aeruginosa Xen 5, we observed a decrease in the tissue uptake of IRDye®800CW 2-deoxyglucose, an indicator of inflammation, and a decrease in bacterial growth from ascitic fluid in mice treated with intravenous recombinant human plasma gelsolin (pGSN) compared to the control vehicle. Pretreatment of the murine macrophage line RAW264.7 with pGSN, followed by addition of Pseudomonas aeruginosa Xen 5, resulted in a dose-dependent increase in the proportion of macrophages with internalized bacteria. This increased uptake was less pronounced when cells were pretreated with pGSN and then centrifuged to remove unbound pGSN before addition of bacteria to macrophages. These observations suggest that recombinant plasma gelsolin can modulate the inflammatory response while at the same time augmenting host antibacterial activity.


Subject(s)
Gelsolin/pharmacology , Inflammation/drug therapy , Phagocytosis/drug effects , Plasma/metabolism , Recombinant Proteins/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Cell Line , Humans , Macrophages/drug effects , Mice , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/drug effects , RAW 264.7 Cells , Sepsis/drug therapy
4.
Oxid Med Cell Longev ; 2020: 4045365, 2020.
Article in English | MEDLINE | ID: mdl-32104532

ABSTRACT

Delineation of factors which affect wound healing would be of immense value to enable on-time or early healing and reduce comorbidities associated with infections or biochemical stress like diabetes. Plasma gelsolin has been identified earlier to significantly enable injury recovery compared to placebo. This study evaluates the role of rhuGSN for its antioxidant and wound healing properties in murine fibroblasts (3T3-L1 cell line). Total antioxidant capacity of rhuGSN increased in a concentration-dependent manner (0.75-200 µg/mL). Cells pretreated with 0.375 and 0.75 µg/mL rhuGSN for 24 h exhibited a significant increase in viability in a MTT assay. Preincubation of cells with rhuGSN for 24 h followed by oxidative stress induced by exposure to H2O2 for 3 h showed cytoprotective effect. rhuGSN at 12.5 and 25 µg/mL concentration showed an enhanced cell migration after 20 h of injury in a scratch wound healing assay. The proinflammatory cytokine IL-6 levels were elevated in the culture supernatant. These results establish an effective role of rhuGSN against oxidative stress induced by H2O2 and in wound healing of 3T3-L1 fibroblast cells.


Subject(s)
Antioxidants/pharmacology , Fibroblasts/drug effects , Gelsolin/pharmacology , Oxidative Stress/drug effects , Wound Healing/drug effects , 3T3-L1 Cells , Animals , Cell Movement/drug effects , Cell Survival/drug effects , Fibroblasts/metabolism , Hydrogen Peroxide/toxicity , Interleukin-6/metabolism , Mice , Reactive Oxygen Species/metabolism
5.
PLoS One ; 14(4): e0215717, 2019.
Article in English | MEDLINE | ID: mdl-31002695

ABSTRACT

The present study provides first evidence on the role of plasma gelsolin in protecting pulmonary thromboembolism and thrombosis in a mouse model. Gelsolin is the most abundant actin depolymerizing protein in plasma and its significantly depleted values have been reported in metabolic disorders including cardiovascular diseases and myocardial infarction. Though gelsolin replacement therapy (GRT) has been shown to be effective in some animal models, no such study has been reported for thrombotic diseases that are acutely in need of bio-therapeutics for immediate and lasting relief. Here, using mice model and recombinant human gelsolin (rhuGSN), we demonstrate the antithrombotic effect of gelsolin in ferric chloride induced thrombosis in carotid artery and thrombin induced acute pulmonary thromboembolism. In thrombosis model, arterial occlusion time was significantly enhanced upon subcutaneous (SC) treatment with 8 mg of gelsolin per mice viz. 15.83 minutes vs. 8 minutes in the placebo group. Pertinently, histopathological examination showed channel formation within the thrombi in the carotid artery following injection of gelsolin. Fluorescence molecular tomography imaging further confirmed that administration of gelsolin reduced thrombus formation following carotid artery injury. In thrombin-induced acute pulmonary thromboembolism, mice pretreated with aspirin or gelsolin showed 100 and 83.33% recovery, respectively. In contrast, complete mortality of mice was observed in vehicle treated group within 5 minutes of thrombin injection. Overall, our studies provide conclusive evidence on the thrombo-protective role of plasma gelsolin in mice model of pulmonary thromboembolism and thrombosis.


Subject(s)
Carotid Artery Thrombosis/prevention & control , Carotid Artery, Common/drug effects , Gelsolin/pharmacology , Pulmonary Embolism/prevention & control , Recombinant Proteins/pharmacology , Thrombosis/prevention & control , Acute Disease , Animals , Carotid Artery Thrombosis/diagnostic imaging , Carotid Artery Thrombosis/physiopathology , Carotid Artery, Common/diagnostic imaging , Carotid Artery, Common/physiopathology , Disease Models, Animal , Female , Fluorescent Dyes/chemistry , Gelsolin/genetics , Humans , Mice, Inbred BALB C , Protective Agents/pharmacology , Pulmonary Embolism/diagnostic imaging , Pulmonary Embolism/physiopathology , Thrombosis/diagnostic imaging , Thrombosis/physiopathology , Tomography/methods
6.
J Nanobiotechnology ; 17(1): 22, 2019 Feb 02.
Article in English | MEDLINE | ID: mdl-30711007

ABSTRACT

BACKGROUND: Human plasma gelsolin (pGSN) is a multifunctional actin-binding protein involved in a variety of biological processes, including neutralization of pro-inflammatory molecules such as lipopolysaccharide (LPS) and lipoteichoic acid (LTA) and modulation of host inflammatory response. It was found that PBP10, a synthetic rhodamine B-conjugated peptide, based on the phosphoinositide-binding site of pGSN, exerts bactericidal activity against Gram-positive and Gram-negative bacteria, interacts specifically with LPS and LTA, and limits microbial-induced inflammatory effects. The therapeutic efficiency of PBP10 when immobilized on the surface of iron oxide-based magnetic nanoparticles was not evaluated, to date. RESULTS: Using the human keratinocyte cell line HaCaT stimulated by bacterially-derived LPS and LTA as an in vitro model of bacterial infection, we examined the anti-inflammatory effects of nanosystems consisting of iron oxide-based magnetic nanoparticles with aminosilane (MNP@NH2) or gold shells (MNP@Au) functionalized by a set of peptides, derived from the phosphatidylinositol 4,5-bisphosphate (PIP2)-binding site of the human plasma protein gelsolin, which also binds LPS and LTA. Our results indicate that these nanosystems can kill both Gram-positive and Gram-negative bacteria and limit the production of inflammatory mediators, including nitric oxide (NO), reactive oxygen species (ROS), and interleukin-8 (IL-8) in the response to heat-killed microbes or extracted bacterial cell wall components. The nanoparticles possess the potential to improve therapeutic efficacy and are characterized by lower toxicity and improved hemocompatibility when compared to free peptides. Atomic force microscopy (AFM) showed that these PBP10-based nanosystems prevented changes in nanomechanical properties of cells that were otherwise stimulated by LPS. CONCLUSIONS: Neutralization of endotoxemia-mediated cellular effects by gelsolin-derived peptides and PBP10-containing nanosystems might be considered as potent therapeutic agents in the improved therapy of bacterial infections and microbial-induced inflammation.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gelsolin/chemistry , Keratinocytes/drug effects , Keratinocytes/immunology , Magnetite Nanoparticles/chemistry , Peptide Fragments/chemistry , Anti-Bacterial Agents/chemistry , Bacteria/drug effects , Binding Sites , Gelsolin/pharmacology , Humans , Inflammation Mediators/metabolism , Keratinocytes/microbiology , Lipopolysaccharides/chemistry , Lipopolysaccharides/toxicity , Peptide Fragments/pharmacology , Peptides/chemistry , Skin Diseases, Bacterial/immunology , Skin Diseases, Bacterial/microbiology , Teichoic Acids/chemistry , Teichoic Acids/toxicity
7.
Am J Physiol Lung Cell Mol Physiol ; 316(1): L105-L113, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30407863

ABSTRACT

We have previously reported that mice genetically deficient in the actin binding protein gelsolin exhibit impaired airway smooth muscle (ASM) relaxation. Primary cultured ASM cells from these mice demonstrate enhanced inositol triphosphate (IP3) synthesis and increased intracellular calcium in response to Gq-coupled agonists. We hypothesized that this was due to increased intracellular availability of unbound phosphatidylinositol 4,5-bisphosphate (PIP2), based on the fact that gelsolin contains a short peptide region that binds PIP2, presumably making it a less available substrate. We now questioned whether a peptide that corresponds to the PIP2 binding region of gelsolin could modulate ASM signaling and contraction. The 10 amino acid sequence of the gelsolin peptide within the PIP2-binding region was incubated with primary cultures of human ASM cells, and IP3 synthesis was measured in response to a Gq-coupled agonist. Gelsolin peptide-treated cells generated less IP3 under basal and bradykinin or acetylcholine (Gq-coupled) conditions. Acetylcholine-induced contractile force measured in isolated tracheal rings from mice and human tracheal muscle strips in organ baths was attenuated in the presence of the gelsolin peptide. The gelsolin peptide also attenuated methacholine-induced airway constriction in murine precision-cut lung slices. Furthermore, this peptide fragment delivered to the respiratory system of mice via nebulization attenuated subsequent methacholine-induced increases in airway resistance in vivo. The current study demonstrates that introduction of this small gelsolin peptide into the airway may be a novel therapeutic option in bronchoconstrictive diseases.


Subject(s)
Bronchoconstriction/drug effects , Gelsolin/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth/metabolism , Peptides/pharmacology , Trachea/metabolism , Animals , Gelsolin/chemistry , Humans , Male , Mice , Muscle, Smooth/pathology , Peptides/chemistry , Phosphatidylinositol 4,5-Diphosphate/metabolism , Trachea/pathology
8.
Stem Cell Res ; 29: 64-75, 2018 05.
Article in English | MEDLINE | ID: mdl-29597129

ABSTRACT

Gelsolin is an actin binding protein present in blood plasma and in cytoplasm of cells including macrophages. Gelsolin has important functions in cell cycle regulation, apoptotic regulation, and morphogenesis. Even though bone marrow macrophages and serum factors are critical for regulating erythropoiesis, the role of gelsolin on human erythroblasts has not been studied. Here, we investigated the effects of human recombinant plasma gelsolin (pGSN) on human immature erythroblasts. CD34+ cells isolated from cord blood were differentiated into erythroid cells in serum-free medium. When pGSN was applied to the culture medium, it accelerated basophilic and polychromatic erythroblast maturation and increased the enucleation rate with highly expressed erythropoiesis-related mRNAs. Also, pGSN was effective in reducing dysplastic changes caused by vincristine, suggesting its role in cell cycle progression at G2/M checkpoints. Also, pGSN activated caspase-3 during maturation stages in which caspase-3 functions as a non-apoptotic maturational signal or a pro-apoptotic signal depending on maturation stages. Our results suggest that pGSN has a pivotal role in maturation of erythroblasts and this factor might be one of the way how bone marrow macrophages and previously unknown serum factors work to control erythropoiesis. pGSN might be used as additive for in vitro production of erythrocytes.


Subject(s)
Erythroblasts/metabolism , Erythrocytes/drug effects , Gelsolin/therapeutic use , Gelsolin/pharmacology , Humans
9.
Environ Toxicol Pharmacol ; 58: 189-195, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29408761

ABSTRACT

Tissues and organs get exposed to high oxygen (O2) supply in hyperoxia conditions. The goal of this research was to investigate the protective effect of actin binding protein gelsolin on hyperoxia-induced hepatotoxicity through histopathology and measurement of oxidative stress parameters and DNA damage in a neonatal Wistar albino rats. The pups were randomly separated to four equal groups such as: normoxia control group (NC), normoxia plus gelsolin group (NG, 10 ng/kg bw/day gelsolin), hyperoxia (≥85% O2) group (HC), hyperoxia plus gelsolin group (HG, ≥85% O2; 10 ng/kg bw/day gelsolin). Histopathological changes of pups in hyperoxia condition were revealed in the form of severe leukocyte infiltration, vascular congestion, necrosis, vacuolar degeneration, binucleated hepatocytes and hemorrhage in the liver tissue. SOD, CAT, GPx and GST activities decreased and MDA level increased in the hyperoxia-induced group in liver tissue (P < 0.05). Tail DNA%, tail length and moment indicating DNA damage statistically increased in hyperoxia treatment groups when compared to controls. Treatment of rats with hyperoxia plus gelsolin prevented hyperoxia-induced changes in tissue structure, antioxidant enzyme activities and MDA level, mean tail DNA% and length. Based on these findings, gelsolin restored these changing to near normal levels but it does not protect completely in the hyperoxia conditions.


Subject(s)
Gelsolin/therapeutic use , Hyperoxia/complications , Liver Diseases/drug therapy , Protective Agents/therapeutic use , Animals , Animals, Newborn , Catalase/metabolism , DNA Damage , Gelsolin/pharmacology , Glutathione Peroxidase/metabolism , Glutathione Transferase/metabolism , Hyperoxia/metabolism , Hyperoxia/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Diseases/etiology , Liver Diseases/metabolism , Liver Diseases/pathology , Malondialdehyde/metabolism , Oxidative Stress/drug effects , Protective Agents/pharmacology , Rats, Wistar , Superoxide Dismutase/metabolism
10.
Sci Rep ; 7(1): 12803, 2017 10 09.
Article in English | MEDLINE | ID: mdl-28993697

ABSTRACT

TNF-α related apoptosis-inducing ligand (TRAIL) selectively kills tumor cells, without damaging normal cells. TRAIL receptors facilitate induction of apoptosis for selective elimination of malignant cells. However, some cancer cells have developed resistances to TRAIL which limits anticancer potential. Gelsolin, a multifunctional actin-binding protein, mediates cell death involving the TRAIL receptors in the hepatic stellate cell line, LX2. Here, we have shown that conditioned medium (CM) containing gelsolin fragments or an N-terminal gelsolin fragment (amino acid residues 1-70) in the presence of TRAIL impairs cell viability of TRAIL resistant transformed human hepatocytes (HepG2). Cell growth regulation by CM and TRAIL was associated with the modulation of p53/Mdm2, Erk and Akt phosphorylation status. The use of N-terminal gelsolin peptide1-70 alone or in combination with TRAIL, induced inhibition of Akt phosphorylation and key survival factors, Mdm2 and Survivin. Treatment of cells with an Akt activator SC79 or p53 siRNA reduced the effects of the N-terminal gelsolin fragment and TRAIL. Together, our study suggests that the N-terminal gelsolin fragment enhances TRAIL-induced loss of cell viability by inhibiting phosphorylation of Akt and promoting p53 function, effecting cell survival.


Subject(s)
Carcinoma, Hepatocellular/pathology , Drug Resistance, Neoplasm/drug effects , Gelsolin/chemistry , Gelsolin/pharmacology , Liver Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Carcinoma, Hepatocellular/enzymology , Cell Death/drug effects , Cell Line, Transformed , Cell Proliferation/drug effects , Cell Survival/drug effects , Culture Media, Conditioned/pharmacology , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver Neoplasms/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , RNA, Small Interfering/metabolism , Survivin/metabolism , Tumor Suppressor Protein p53/metabolism
11.
J Med Chem ; 60(16): 6991-6997, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28700225

ABSTRACT

Structural optimization of a peptidomimetic antagonist of formyl peptide receptor 2 (FPR2) was explored by an approach involving combination of elements from the two most potent FPR2 antagonists described: a Rhodamine B-conjugated 10-residue gelsonin-derived peptide (i.e., PBP10, RhB-QRLFQVKGRR-OH) and the palmitoylated α-peptide/ß-peptoid hybrid Pam-(Lys-ßNspe)6-NH2. This generated an array of hybrid compounds from which a new subclass of receptor-selective antagonists was identified. The most potent representatives displayed activity in the low nanomolar range. The resulting stable and potent FPR2-selective antagonists (i.e., RhB-(Lys-ßNphe)n-NH2; n = 4-6) are expected to become valuable tools in further elucidation of the physiological role of FPR2 in health and disease.


Subject(s)
Gelsolin/pharmacology , Peptide Fragments/pharmacology , Peptides/pharmacology , Peptidomimetics/pharmacology , Receptors, Formyl Peptide/antagonists & inhibitors , Receptors, Lipoxin/antagonists & inhibitors , Enzyme Activators/pharmacology , Gelsolin/chemical synthesis , Humans , Molecular Structure , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , NADPH Oxidases/metabolism , Neutrophils/metabolism , Oligopeptides/pharmacology , Peptide Fragments/chemical synthesis , Peptides/chemical synthesis , Peptides/chemistry , Peptidomimetics/chemical synthesis , Peptidomimetics/chemistry , Signal Transduction/drug effects , Structure-Activity Relationship , Superoxides/analysis
12.
Pregnancy Hypertens ; 6(4): 333-339, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27939478

ABSTRACT

BACKGROUND: Preeclampsia, a pregnancy-specific inflammatory disorder, is characterized by high levels of anti-angiogenic protein, soluble fms-like tyrosine kinase 1 (sFlt1), in the maternal circulation. sFlt1 producing molecular machinery is present in syncytiotrophoblast extracellular vesicles that are released by the placenta into maternal plasma during normal pregnancy, a process greatly accelerated in preeclampsia. We hypothesized that syncytiotrophoblast extracellular vesicles exposes cytoplasmic actin to plasma resulting in depletion of plasma gelsolin (pGSN), an abundant plasma protein that scavenges circulating actin and other pro-inflammatory mediators. OBJECTIVE: To test whether pGSN levels would be lower in preeclampsia and to assess whether recombinant human plasma gelsolin (rhpGSN) may promote placental health by decreasing shedding of syncytiotrophoblast extracellular vesicles. METHODS: We tested pGSN levels in third trimester plasma samples from women with preeclampsia and non-hypertensive pregnancies. We then assessed whether rhpGSN may act as a negative regulator of syncytial shedding in placental explant culture and dynamic mechanical stretch studies. RESULTS: pGSN levels fall in late pregnancy and decline further in preeclampsia patients. Recombinant human pGSN (rhpGSN) at 100µg/ml limits spontaneous syncytiotrophoblast vesicle release and sFlt1 protein dissemination by normal placental explants. Higher rhpGSN doses (500µg/ml) also limit syncytiotrophoblast vesicle and sFlt1 dissemination from preeclamptic placental explants. rhpGSN also mitigates syncytiotrophoblast vesicle during dynamic mechanical stretch. CONCLUSIONS: 1) pGSN, an anti-inflammatory factor of maternal origin is reduced in preeclampsia and may contribute to disease progression and 2) exogenous rhpGSN supplementation can limit the dissemination of toxic syncytiotrophoblast vesicle that characterizes the disease state.


Subject(s)
Extracellular Vesicles , Gelsolin/blood , Gelsolin/pharmacology , Pre-Eclampsia/blood , Recombinant Proteins/pharmacology , Trophoblasts/drug effects , Adult , Case-Control Studies , Female , Humans , Pregnancy , Pregnancy Trimester, Third , Tissue Culture Techniques , Trophoblasts/physiology , Vascular Endothelial Growth Factor Receptor-1/metabolism , Young Adult
13.
Dev Biol ; 415(1): 64-74, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27178669

ABSTRACT

In order to interact with the egg and undergo acrosomal exocytosis or the acrosome reaction (AR), mammalian spermatozoa must undergo a series of biochemical changes in the female reproductive tract, collectively called capacitation. We showed that F-actin is formed during sperm capacitation and fast depolymerization occurs prior to the AR. We hypothesized that F-actin protects the sperm from undergoing spontaneous-AR (sAR) which decreases fertilization rate. We show that activation of the actin-severing protein gelsolin induces a significant increase in sAR. Moreover, inhibition of CaMKII or PLD during sperm capacitation, caused an increase in sAR and inhibition of F-actin formation. Spermine, which leads to PLD activation, was able to reverse the effects of CaMKII inhibition on sAR-increase and F-actin-decrease. Furthermore, the increase in sAR and the decrease in F-actin caused by the inactivation of the PLD-pathway, were reversed by activation of CaMKII using H2O2 or by inhibiting protein phosphatase 1 which enhance the phosphorylation and oxidation states of CaMKII. These results indicate that two distinct pathways lead to F-actin formation in the sperm capacitation process which prevents the occurrence of sAR.


Subject(s)
Acrosome Reaction/physiology , Acrosome/enzymology , Actins/metabolism , Actin Cytoskeleton/ultrastructure , Animals , Calcimycin/pharmacology , Calcium Signaling/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Cattle , Enzyme Activation/drug effects , Exocytosis/physiology , Gelsolin/metabolism , Gelsolin/pharmacology , Hydrogen Peroxide/pharmacology , Male , Marine Toxins , Oxazoles/pharmacology , Peptide Fragments/pharmacology , Phospholipase D/antagonists & inhibitors , Phospholipase D/physiology , Polymerization , Sperm Capacitation/physiology , Spermatozoa/drug effects , Spermatozoa/ultrastructure
14.
Life Sci ; 155: 1-9, 2016 Jun 15.
Article in English | MEDLINE | ID: mdl-27157519

ABSTRACT

AIMS: Deaths associated with cancer metastasis have steadily increased making the need for newer, anti-metastatic therapeutics imparative. Gelsolin and vimentin, actin binding proteins expressed in metastatic tumors, participate in actin remodelling and regulate cell migration. Docosahexaenoic acid (DHA) limits cancer cell proliferation and adhesion but the mechanisms involved in reducing metastatic phenotypes are unknown. We aimed to investigate the effects of DHA on gelsolin and vimentin expression, and ultimately cell migration and proliferation, in this context. MAIN METHODS: Non-invasive lung epithelial cells (MLE12) and invasive lung cancer cells (A549) were treated with DHA (30µmol/ml) or/and 8 bromo-cyclic adenosine monophosphate (8 Br-cAMP) (300µmol/ml) for 6 or 24h either before (pre-treatment) or after (post-treatment) plating in transwells. Migration was assessed by the number of cells that progressed through the transwell. Gelsolin and vimentin expression were measured by Western blot and confocal microscopy in cells, and by immunohistochemistry in human lung cancer biopsy samples. KEY FINDINGS: A significant decrease in cell migration was detected for A549 cells treated with DHA verses control but this same decrease was not seen in MLE12 cells. DHA and 8 Br-cAMP altered gelsolin and vimentin expression but no clear pattern of change was observed. Immunofluorescence staining indicated slightly higher vimentin expression in human lung tissue that was malignant compared to control. SIGNIFICANCE: Collectively, our data indicate that DHA inhibits cancer cell migration and further suggests that vimentin and gelsolin may play secondary roles in cancer cell migration and proliferation, but are not the primary regulators.


Subject(s)
Cell Movement/drug effects , Docosahexaenoic Acids/pharmacology , Gelsolin/pharmacology , Lung Neoplasms/pathology , Vimentin/metabolism , A549 Cells , Humans , Lung Neoplasms/metabolism
15.
Tumour Biol ; 37(6): 7165-74, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26662962

ABSTRACT

The gelsolin (GSN) has been involved in the regulation of tumor formation and development, but the biological role of GSN in the pathogenesis of osteosarcoma (OS) has not been well characterized. In this study, we show that high expression of GSN was observed in OS tissues and correlated with tumor size, advanced Enneking stage, and poor patient prognosis. Knockdown of GSN significantly inhibited cell proliferation and invasiveness in the OS cell lines. Furthermore, stable overexpression of GSN in OS cells resulted in a significant increase in cell growth and motility with the downregulation of p-AKT and p-P38 pathway. Finally, overexpression of GSN promoted growth of OS tumors in SCID mice. Taken together, these results provided the first evidence that GSN might contribute to OS cancer development and could be an attractive therapeutic target for patients with OS.


Subject(s)
Cell Movement/drug effects , Cell Proliferation/drug effects , Gelsolin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Osteosarcoma/pathology , Proto-Oncogene Proteins c-akt/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Adolescent , Animals , Apoptosis/drug effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Staging , Osteosarcoma/drug therapy , Osteosarcoma/metabolism , Prognosis , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/genetics
16.
PLoS One ; 10(9): e0135558, 2015.
Article in English | MEDLINE | ID: mdl-26426535

ABSTRACT

Plasma gelsolin levels significantly decline in several disease conditions, since gelsolin gets scavenged when it depolymerizes and caps filamentous actin released in the circulation following tissue injury. It is well established that our body require/implement inflammatory and analgesic responses to protect against cell damage and injury to the tissue. This study was envisaged to examine analgesic and anti-inflammatory activity of exogenous gelsolin (8 mg/mouse) in mice models of pain and acute inflammation. Administration of gelsolin in acetic acid-induced writhing and tail immersion tests not only demonstrated a significant reduction in the number of acetic acid-induced writhing effects, but also exhibited an analgesic activity in tail immersion test in mice as compared to placebo treated mice. Additionally, anti-inflammatory function of gelsolin (8 mg/mouse) compared with anti-inflammatory drug diclofenac sodium (10 mg/kg)] was confirmed in the carrageenan injection induced paw edema where latter was measured by vernier caliper and fluorescent tomography imaging. Interestingly, results showed that plasma gelsolin was capable of reducing severity of inflammation in mice comparable to diclofenac sodium. Analysis of cytokines and histopathological examinations of tissue revealed administration of gelsolin and diclofenac sodium significantly reduced production of pro-inflammatory cytokines, TNF-α and IL-6. Additionally, carrageenan groups pretreated with diclofenac sodium or gelsolin showed a marked decrease in edema and infiltration of inflammatory cells in paw tissue. Our study provides evidence that administration of gelsolin can effectively reduce the pain and inflammation in mice model.


Subject(s)
Analgesics/pharmacology , Anti-Inflammatory Agents/pharmacology , Carrageenan/metabolism , Edema/pathology , Gelsolin/pharmacology , Inflammation/drug therapy , Acetic Acid/adverse effects , Animals , Diclofenac/pharmacology , Disease Models, Animal , Extremities/pathology , Female , Mice, Inbred BALB C , Tail/drug effects
17.
Am J Physiol Lung Cell Mol Physiol ; 309(1): L11-6, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25957291

ABSTRACT

Plasma gelsolin (pGSN) functions as part of the "extracellular actin-scavenging system," but its potential to improve host defense against infection has not been studied. In a mouse model of primary pneumococcal pneumonia, recombinant human pGSN (rhu-pGSN) caused enhanced bacterial clearance, reduced acute inflammation, and improved survival. In vitro, rhu-pGSN rapidly improved lung macrophage uptake and killing of bacteria (Streptococcus pneumoniae, Escherichia coli, and Francisella tularensis). pGSN triggers activating phosphorylation (Ser(1177)) of macrophage nitric oxide synthase type III (NOS3), an enzyme with important bactericidal functions in lung macrophages. rhu-pGSN failed to enhance bacterial killing by NOS3(-/-) macrophages in vitro or bacterial clearance in NOS3(-/-) mice in vivo. Prophylaxis with immunomodulators may be especially relevant for patients at risk for secondary bacterial pneumonia, e.g., after influenza. Treatment of mice with pGSN challenged with pneumococci on postinfluenza day 7 (the peak of enhanced susceptibility to secondary infection) caused a ∼15-fold improvement in bacterial clearance, reduced acute neutrophilic inflammation, and markedly improved survival, even without antibiotic therapy. pGSN is a potential immunomodulator for improving lung host defense against primary and secondary bacterial pneumonia.


Subject(s)
Gelsolin/pharmacology , Lung/microbiology , Macrophages, Alveolar/immunology , Nitric Oxide Synthase Type III/immunology , Pneumonia, Pneumococcal/immunology , Animals , Cell Line , Disease Models, Animal , Disease Susceptibility , Escherichia coli/immunology , Francisella tularensis/immunology , Gelsolin/blood , Inflammation/drug therapy , Inflammation/immunology , Influenza A Virus, H1N1 Subtype/immunology , Macrophages, Alveolar/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type III/genetics , Orthomyxoviridae Infections/immunology , Phagocytosis/immunology , Pneumonia, Pneumococcal/mortality , Pneumonia, Pneumococcal/prevention & control , Recombinant Proteins/pharmacology , Streptococcus pneumoniae/immunology
18.
J Cyst Fibros ; 14(5): 587-93, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25682700

ABSTRACT

BACKGROUND: DNase (Pulmozyme) effectiveness in cystic fibrosis treatment is in some cases limited by its inability to access DNA trapped within bundles in highly viscous fluids that also contain actin. Dissociating DNA-containing bundles using actin depolymerizing agents and polyanions has potential to increase DNase efficacy. METHODS: Fluorescence measurements of YOYO-1 and a rheological creep-recovery test quantified DNA content and viscoelasticity in 150 sputum samples from adult CF patients and their susceptibility to fluidization by DNase1, alone and in combination with gelsolin and poly-aspartate (p-Asp). Fluidization of sputum by these agents is compared to their capacity to increase antibacterial activity in sputum, measured using a luminescent Pseudomonas aeruginosa strain and a bacterial killing assay. RESULTS: The polyanion p-Asp (1-50 µg/g of sputum), the actin severing protein gelsolin (10-90 µg/g) and their combination enhance the ability of DNase 1 to increase the abnormally low mechanical compliance of CF sputum and to promote bacterial killing in sputum by colistin and tobramycin, two antibiotics commonly used to treat CF. CONCLUSIONS: Addition of low concentrations of p-ASP or gelsolin can increase the therapeutic value of Pulmozyme (DNase 1).


Subject(s)
Cystic Fibrosis/metabolism , Deoxyribonuclease I/pharmacology , Gelsolin/pharmacology , Peptides/pharmacology , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/drug effects , Sputum/chemistry , Adolescent , Adult , Child , Compliance , Cystic Fibrosis/drug therapy , Cystic Fibrosis/microbiology , Deoxyribonuclease I/metabolism , Female , Humans , Male , Middle Aged , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/isolation & purification , Recombinant Proteins/pharmacology , Suppuration/microbiology , Young Adult
19.
Cell Biochem Biophys ; 71(1): 389-96, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25164111

ABSTRACT

The present study was conducted to explore the protective effect of exogenous gelsolin (GSN) in mice exposed to high-dose of radiation. Changes in the levels of GSNs in peripheral blood of mice and cytoplasm of cultured human intestinal epithelial cells (HIECs) were analyzed after their exposure to different doses of (137)Cs γ-rays at a fixed dose rate. The coagulation associated indices, such as prothrombin time (PT) and activated partial thromboplastin time (APTT) were measured. Effect on radiation-mediated oxidative damage was evaluated by estimating the altered glutathione (GSH) and malondialdehyde (MDA) concentrations in the blood. The results showed that radiation induced a pronounced decrease in the pGSN blood levels. However, the cGSN levels of irradiated HIECs were increased in a dose-dependent manner. Administration of recombinant human pGSN to irradiated mice resulted in an ameliorated clotting time as indicated by the PT and the APTT indices. The treatment of mice with hpGSN enhanced the blood levels of GSH while MDA concentrations were decreased indicating an improved antioxidant status. These results suggest that GSNs might play a regulatory role in the suppression of the tissue damage induced by acute radiation exposure.


Subject(s)
Gelsolin/pharmacology , Radiation Injuries/drug therapy , Animals , Blood Coagulation/drug effects , Blood Coagulation/radiation effects , Catalase/blood , Cytoplasm/drug effects , Cytoplasm/metabolism , Dose-Response Relationship, Radiation , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/radiation effects , Gamma Rays/adverse effects , Gelsolin/blood , Gelsolin/metabolism , Gelsolin/therapeutic use , Glutathione/blood , Humans , Intestines/cytology , Male , Malondialdehyde/blood , Mice , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Partial Thromboplastin Time , Prothrombin Time , Radiation Injuries/blood , Radiation Injuries/metabolism , Radiation Injuries/physiopathology , Superoxide Dismutase/metabolism
20.
J Diabetes Res ; 2014: 152075, 2014.
Article in English | MEDLINE | ID: mdl-25478578

ABSTRACT

The study aims to map plasma gelsolin (pGSN) levels in diabetic humans and mice models of type II diabetes and to evaluate the efficacy of gelsolin therapy in improvement of diabetes in mice. We report that pGSN values decrease by a factor of 0.45 to 0.5 in the blood of type II diabetic humans and mice models. Oral glucose tolerance test in mice models showed that subcutaneous administration of recombinant pGSN and its F-actin depolymerizing competent versions brought down blood sugar levels comparable to Sitagliptin, a drug used to manage hyperglycemic condition. Further, daily dose of pGSN or its truncated versions to diabetic mice for a week kept sugar levels close to normal values. Also, diabetic mice treated with Sitagliptin for 7 days, showed increase in their pGSN values with the decrease in blood glucose as compared to their levels at the start of treatment. Gelsolin helped in improving glycemic control in diabetic mice. We propose that gelsolin level monitoring and replacement of F-actin severing capable gelsolin(s) should be considered in diabetic care.


Subject(s)
Actins/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Gelsolin/blood , Gelsolin/pharmacology , Hypoglycemic Agents/pharmacology , Peptide Fragments/pharmacology , Adult , Animals , Biomarkers/blood , Blood Glucose/drug effects , Blood Glucose/metabolism , Case-Control Studies , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/diagnosis , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/genetics , Diet, High-Fat , Down-Regulation , Female , Humans , Male , Mice , Mice, Inbred C57BL , Polymerization , Pyrazines/pharmacology , Recombinant Proteins/pharmacology , Sitagliptin Phosphate , Streptozocin , Time Factors , Triazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...