Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
Opt Express ; 31(26): 43722-43731, 2023 Dec 18.
Article in English | MEDLINE | ID: mdl-38178462

ABSTRACT

Enhancing the ability to resolve axial details is crucial in three-dimensional optical imaging. We provide experimental evidence showcasing the ultimate precision achievable in axial localization using vortex beams. For Laguerre-Gauss (LG) beams, this remarkable limit can be attained with just a single intensity scan. This proof-of-principle demonstrates that microscopy techniques based on LG vortex beams can potentially benefit from the introduced quantum-inspired superresolution protocol.

3.
Ann Rheum Dis ; 76(3): 612-619, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27597652

ABSTRACT

OBJECTIVES: Tristetraprolin (TTP), a negative regulator of many pro-inflammatory genes, is strongly expressed in rheumatoid synovial cells. The mitogen-activated protein kinase (MAPK) p38 pathway mediates the inactivation of TTP via phosphorylation of two serine residues. We wished to test the hypothesis that these phosphorylations contribute to the development of inflammatory arthritis, and that, conversely, joint inflammation may be inhibited by promoting the dephosphorylation and activation of TTP. METHODS: The expression of TTP and its relationship with MAPK p38 activity were examined in non-inflamed and rheumatoid arthritis (RA) synovial tissue. Experimental arthritis was induced in a genetically modified mouse strain, in which endogenous TTP cannot be phosphorylated and inactivated. In vitro and in vivo experiments were performed to test anti-inflammatory effects of compounds that activate the protein phosphatase 2A (PP2A) and promote dephosphorylation of TTP. RESULTS: TTP expression was significantly higher in RA than non-inflamed synovium, detected in macrophages, vascular endothelial cells and some fibroblasts and co-localised with MAPK p38 activation. Substitution of TTP phosphorylation sites conferred dramatic protection against inflammatory arthritis in mice. Two distinct PP2A agonists also reduced inflammation and prevented bone erosion. In vitro anti-inflammatory effects of PP2A agonism were mediated by TTP activation. CONCLUSIONS: The phosphorylation state of TTP is a critical determinant of inflammatory responses, and a tractable target for novel anti-inflammatory treatments.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/enzymology , Protein Phosphatase 2/metabolism , Tristetraprolin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Amino Alcohols/therapeutic use , Animals , Apolipoproteins E/therapeutic use , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/prevention & control , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Enzyme Activation/drug effects , Fibroblasts/metabolism , Humans , MAP Kinase Signaling System , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy , Phosphorylation , Protein Phosphatase 2/drug effects , RNA, Messenger/metabolism , Serine/metabolism , Synovial Membrane/metabolism , Tristetraprolin/genetics
6.
Biomed Khim ; 60(4): 515-21, 2014.
Article in Russian | MEDLINE | ID: mdl-25249536

ABSTRACT

The neuroprotective activity of apolipoprotein E (apoE) peptide mimetic Cog1410, containing amino acid sequence of the receptor-binding domain apoE, has been investigated in transgenic lines of Drosophila melanogaster expressing human APP and beta-secretase. Expression of two transgenes caused neuropathological processes attributed to Alzheimer's disease: neurodegeneration, cognitive abnormality and amyloid deposits formation in brain. It was shown that Cog 1410 reduces neurodegeneration in brain of transgenic flies and improves cognitive functions (odor recognition). These data suggest that Cog1410 is a potential neuroprotector that can be used in AD treatment.


Subject(s)
Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/genetics , Apolipoproteins E/pharmacology , Cognition/drug effects , Drosophila melanogaster/drug effects , Neuroprotective Agents/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Animals, Genetically Modified , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Brain/pathology , Disease Models, Animal , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression , Humans , Odorants , Olfactory Perception/drug effects , Transgenes
8.
Neurodegener Dis ; 10(1-4): 122-6, 2012.
Article in English | MEDLINE | ID: mdl-22326991

ABSTRACT

BACKGROUND: After age, the second largest risk factor for Alzheimer's disease (AD) is apolipoprotein E (APOE) genotype, where APOE4 is associated with lower apoE protein levels, more severer brain pathology, enhanced inflammation and disease. Small peptides corresponding to the receptor-binding region of apoE mimic the anti-inflammatory activity of the apoE holoprotein. These apoE mimetics greatly improve behavioral outcomes and neuronal survival in head trauma models that display AD pathology and neuronal loss. OBJECTIVE: To determine whether apoE mimetics change behavior, inflammation and pathology in CVND-AD (SwDI-APP/NOS2(-/-)) transgenic mice. METHODS: Starting at 9 months, apoE peptides were subcutaneously administered 3 times per week for 3 months followed by behavioral, histochemical and biochemical testing. RESULTS: Treatment with apoE mimetics significantly improved behavior while decreasing the inflammatory cytokine IL-6, neurofibrillary tangle-like and amyloid plaque-like structures. Biochemical measures matched the visible pathological results. CONCLUSIONS: Treatment with apoE mimetics significantly improved behavior, reduced inflammation and reduced pathology in CVND-AD mice. These improvements are associated with apoE-mimetic-mediated increases in protein phosphatase 2A activity. Testing in additional AD models showed similar benefits, reinforcing this novel mechanism of action of apoE mimetics. These data suggest that the combination of anti-inflammatory and neuroprotective activities of apoE mimetics represents a new generation of potential therapeutics for AD.


Subject(s)
Alzheimer Disease/complications , Alzheimer Disease/pathology , Apolipoproteins E/therapeutic use , Behavioral Symptoms/drug therapy , Brain/metabolism , Neurofibrillary Tangles/drug effects , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Behavioral Symptoms/etiology , Brain/drug effects , Brain/pathology , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Maze Learning/drug effects , Mice , Mice, Transgenic , Motor Activity/drug effects , Mutation/genetics , Nitric Oxide Synthase Type II/deficiency , Phosphopyruvate Hydratase/metabolism , RNA, Messenger/metabolism , Time Factors , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
9.
Oncogene ; 30(22): 2504-13, 2011 Jun 02.
Article in English | MEDLINE | ID: mdl-21297667

ABSTRACT

The SET oncoprotein participates in cancer progression by affecting multiple cellular processes, inhibiting the tumor suppressor protein phosphatase 2A (PP2A), and inhibiting the metastasis suppressor nm23-H1. On the basis of these multiple activities, we hypothesized that targeted inhibition of SET would have multiple discrete and measurable effects on cancer cells. Here, the effects of inhibiting SET oncoprotein function on intracellular signaling and proliferation of human cancer cell lines was investigated. We observed the effects of COG112, a novel SET interacting peptide, on PP2A activity, Akt signaling, nm23-H1 activity and cellular migration/invasion in human U87 glioblastoma and MDA-MB-231 breast adenocarcinoma cancer cell lines. We found that COG112 interacted with SET protein and inhibited the association between SET and PP2A catalytic subunit (PP2A-c) and nm23-H1. The interaction between COG112 and SET caused PP2A phosphatase and nm23-H1 exonuclease activities to increase. COG112-mediated increases in PP2A activity resulted in the inhibition of Akt signaling and cellular proliferation. Additionally, COG112 inhibited SET association with Ras-related C(3) botulinum toxin substrate 1 (Rac1), leading to decreased cellular migration and invasion. COG112 treatment releases the SET-mediated inhibition of the tumor suppressor PP2A, as well as the metastasis suppressor nm23-H1. These results establish SET as a novel molecular target and that the inhibition of SET may have beneficial effects in cancer chemotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Histone Chaperones/antagonists & inhibitors , Neoplasms/drug therapy , Peptides/therapeutic use , Transcription Factors/antagonists & inhibitors , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , DNA-Binding Proteins , Humans , NM23 Nucleoside Diphosphate Kinases/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , rac1 GTP-Binding Protein/metabolism
10.
Tsitologiia ; 53(12): 959-67, 2011.
Article in Russian | MEDLINE | ID: mdl-22359955

ABSTRACT

One of the earliest neuropathological symptoms of Alzheimer's disease is the loss of synapses, which preceed the formation of amyloidosis and neurodegeneration. Although most cases of early-onset familial Alzheimer's disease are caused by mutations in the presenilin 1 (PS1) gene, the functions of PS1 and its role in synaptic disfunction are not yet completely understood. In this paper we analysed of the intracellular and extracellular distribution of PS1 in the cultures of mouse cortical embryonic neurons. We found that PS1 is concentrated on the surface of the growth cone and at neurite contact sites. PS1 was also found in synapses where it is co-localized with synaptophysin. Independent evidense of involvement of PS1 in synaptic function we obtained by transfection of neurons with GFP-PS1 cDNA. GFP was colocalized with synaptophysin in transfected cultures. GFP-immunoprecepitates from transfected neurons contained processed N-cadherin. This result presents an additional proof of involvment PS1 in synapse formation. To evaluate the role of PS1 inactivation in the synaptic functions, we compare synaptic density in neuronal cell cultures from PS1 knockout mice PS1 (-/-) and wild type mice PS1 (+/+). Our results clearly show that PS1 (-/-) displayed a low number of morphological synapses in comparing with wild type culture PS1 (+/+). In summary, our results indicate a role of PS1 in synaptic function.


Subject(s)
Cerebral Cortex/metabolism , Embryo, Mammalian/metabolism , Growth Cones/metabolism , Neurites/metabolism , Presenilin-1/metabolism , Synapses/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Cells, Cultured , Cerebral Cortex/cytology , Embryo, Mammalian/cytology , Humans , Mice , Mice, Knockout , Presenilin-1/genetics , Synapses/genetics , Synaptophysin , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
11.
Biofizika ; 55(5): 862-7, 2010.
Article in Russian | MEDLINE | ID: mdl-21033353

ABSTRACT

Experimental evidence has been obtained that mutations in the presenilin 1 (PS1) gene in familial Alzheimer's disease can lead to the disturbance of cell adhesion in model cell cultures. It was shown that, in L fibroblasts of mice with stable expression of GFP-PS1 cDNA containing G209V or E319G mutations, cell-cell interactions and the accumulation of GFP-PS1 cDNA in intercellular contacts are disturbed. Similar results were obtained in transfected human epithelial Hep2 cells. It is assumed that mutations in familial Alzheimer's disease lead to the disturbance of the functions of presenelin 1 in cell adhesion.


Subject(s)
Alzheimer Disease/pathology , Presenilin-1/genetics , Alzheimer Disease/genetics , Animals , Cell Adhesion , Cell Line , Epithelial Cells/physiology , Fibroblasts/physiology , Humans , Mice , Mutation
12.
Euro Surveill ; 15(20)2010 May 20.
Article in English | MEDLINE | ID: mdl-20504390

ABSTRACT

After ten years of being measles free, Slovenia experienced a cluster with secondary transmission in a hospital setting in March 2010. The index case, a resident of Ireland, was hospitalised on the day after his arrival to Slovenia and diagnosed with measles two days later. After his discharge, two cases of measles were notified, a hospital staff member and a visitor to the clinic, suggesting transmission in a hospital setting.


Subject(s)
Cross Infection/etiology , Hospitals , Measles/transmission , Adolescent , Adult , Child , Child, Preschool , Humans , Infant , Measles/epidemiology , Morbillivirus/isolation & purification , Slovenia/epidemiology , Travel , Young Adult
13.
Neuroscience ; 169(1): 516-31, 2010 Aug 11.
Article in English | MEDLINE | ID: mdl-20434528

ABSTRACT

A central issue in the pathogenesis of tauopathy is the question of how tau protein dysfunction leads to neurodegeneration. We have previously demonstrated that the absence of tau protein is associated with destabilization of microtubules and impaired neurite outgrowth (Dawson et al., 2001; Rapoport et al., 2002). We now hypothesize that the absence of functional tau protein may render the central nervous system more vulnerable to secondary insults such as the overexpression of mutated beta amyloid precursor protein (APP) and traumatic brain injury. We therefore crossed tau knockout mice (Dawson et al., 2001) to mice overexpressing a mutated human APP (APP(670,671), A(sw)) (Hsiao et al., 1996) and created a mouse model (A(sw)/mTau(-/-)) that provides evidence that the loss of tau function causes degeneration of neuronal processes. The overexpression of APP(670,671) in tau knockout mice, elicits the extensive formation of axonal spheroids. While spheroids are only found associated with Abeta plaques in mice expressing APP(670,671) on an endogenous mouse tau background (Irizarry et al., 1997), A(sw)/mTau(-/-) mice have spheroids not only surrounding Abeta plaques but also in white matter tracks and in the neuropil. Plaque associated and neuropil dystrophic neurites and spheroids are prominent features of Alzheimer's disease (Masliah et al., 1993; Terry, 1996; Stokin et al., 2005), and our current data suggests that loss of tau function may lead to neurodegeneration.


Subject(s)
Alzheimer Disease/pathology , Axons/pathology , Nerve Degeneration/pathology , tau Proteins/deficiency , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/immunology , Amyloid beta-Protein Precursor/toxicity , Animals , Ataxia/etiology , Brain Injuries/genetics , Brain Injuries/metabolism , Brain Injuries/pathology , Disease Models, Animal , Humans , Immunization , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Electron , Neurites/ultrastructure , Neuropil/ultrastructure , Plaque, Amyloid/ultrastructure , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/toxicity , tau Proteins/genetics , tau Proteins/physiology
14.
Oncogene ; 28(43): 3837-46, 2009 Oct 29.
Article in English | MEDLINE | ID: mdl-19701246

ABSTRACT

The chemopreventative effects of dithiolethione compounds are attributed to their activation of antioxidant response elements (AREs) by reacting with the Nrf2/Keap1 protein complex. In this study, we show antiproliferative effects of the dithiolethione compound ACS-1 in human cancer cell lines (A549 and MDA-MB-231) by increasing the activity of the tumor suppressor protein phoshatase 2A (PP2A). ACS-1 inhibited epidermal growth factor (EGF)-induced cellular proliferation in a concentration- and time-dependent manner. Akt activation, as determined by serine-473 phosphorylation, was inhibited by ACS-1 in cells stimulated with either EGF or fibronectin. Furthermore, ACS-1 inhibited mammalian target of rapamycin signaling and decreased c-myc protein levels. ACS-1 did not proximally alter EGF receptor or integrin signaling, but caused a concentration-dependent increase in PP2A activity. The effect of ACS-1 on Akt activation was not observed in the presence of the PP2A inhibitor okadaic acid. ACS-1 effects on PP2A activity were independent of ARE activation and cAMP formation. In addition to ACS-1, other dithiolethione compounds showed similar effects in reducing Akt activation, suggesting that this class of compounds may have other effects beyond chemoprevention.


Subject(s)
Anticarcinogenic Agents/pharmacology , Breast Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Lung Neoplasms/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
15.
Biomed Khim ; 55(1): 41-9, 2009.
Article in Russian | MEDLINE | ID: mdl-19351032

ABSTRACT

Protein transduction domain (PTD)-peptides greatly facilitate the delivery of high molecular weight macromolecules across the blood-brain barrier (BBB). This BBB-transport function is highly desirable and helps to enable the development of new therapeutics for treatment of brain disorders. However, the drug discovery process is limited by the generation of a simple and reliable BBB model that is amenable to testing of large number of samples and simultaneously, reproduces the physiological and functional characteristics of the human BBB. To address these challenges, we have studied whether the PTD-peptide penetratin, derived from a Drosophila Antennapedia homeodomain protein, is capable of crossing the BBB in Drosophila while carrying a cargo into the fly brain. An initial in vivo experiment in Drosophila showed that abdominal injection of biotin-tagged penetratin permeated the BBB. The same effect was observed for biotin-tagged penetratin fused with apoE mimetic peptide with demonstrated anti-inflammatory and neuroprotective activities.


Subject(s)
Antennapedia Homeodomain Protein/pharmacology , Blood-Brain Barrier , Brain Diseases/drug therapy , Carrier Proteins/pharmacology , Drosophila Proteins/pharmacology , Peptides/pharmacology , Animals , Cell-Penetrating Peptides , Drosophila melanogaster , Drug Delivery Systems , Humans , Protein Structure, Tertiary
16.
Neuroscience ; 159(3): 1055-69, 2009 Mar 31.
Article in English | MEDLINE | ID: mdl-19356689

ABSTRACT

The neurovascular unit (NVU) comprises cerebral blood vessels and surrounding astrocytes, neurons, perivascular microglia and pericytes. Astrocytes associated with the NVU are responsible for maintaining cerebral blood flow and ionic and osmotic balances in the brain. A significant proportion of individuals with Alzheimer's disease (AD) have vascular amyloid deposits (cerebral amyloid angiopathy, CAA) that contribute to the heterogeneous nature of the disease. To determine whether NVU astrocytes are affected by the accumulation of amyloid at cerebral blood vessels we examined astrocytic markers in four transgenic mouse models of amyloid deposition. These mouse models represent mild CAA, moderate CAA with disease progression to tau pathology and neuron loss, severe CAA and severe CAA with disease progression to tau pathology and neuron loss. We found that CAA and disease progression both resulted in distinct NVU astrocytic changes. CAA causes a loss of apparent glial fibrillary acidic protein (GFAP)-positive astrocytic end-feet and loss of water channels (aquaporin 4) localized to astrocytic end feet. The potassium channels Kir4.1, an inward rectifying potassium channel, and BK, a calcium-sensitive large-conductance potassium channel, were also lost. The anchoring protein, dystrophin 1, is common to these channels and was reduced in association with CAA. Disease progression was associated with a phenotypic switch in astrocytes indicated by a loss of GFAP-positive cells and a gain of S100 beta-positive cells. Aquaporin 4, Kir4.1 and dystrophin 1 were also reduced in autopsied brain tissue from individuals with AD that also display moderate and severe CAA. Together, these data suggest that damage to the neurovascular unit may be a factor in the pathogenesis of Alzheimer's disease.


Subject(s)
Alzheimer Disease/physiopathology , Aquaporin 4/metabolism , Astrocytes/physiology , Cerebral Amyloid Angiopathy/physiopathology , Potassium Channels/metabolism , Aged, 80 and over , Amyloid beta-Protein Precursor/genetics , Animals , Astrocytes/pathology , Brain/blood supply , Brain/physiopathology , Disease Models, Animal , Dystrophin/metabolism , Female , Glial Fibrillary Acidic Protein/metabolism , Humans , Kv1.6 Potassium Channel , Male , Mice , Mice, Transgenic , Nitric Oxide Synthase Type II/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Protease Nexins , RNA, Messenger , Receptors, Cell Surface/genetics , Shaker Superfamily of Potassium Channels/metabolism
17.
Genetika ; 45(1): 119-26, 2009 Jan.
Article in Russian | MEDLINE | ID: mdl-19239106

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disease whose main pathomorphological sign is synapse degeneration in the cortex and hippocampus. Abnormal synaptogenesis precedes amyloidosis and neurodegeneration and correlates with memory impairment during the early clinical phase. Mutations in the amyloid precursor protein (APP) gene cause familial AD and enhance the secretion of amyloid-beta-protein (Abeta). However, it remains unclear in what way APP and Abeta are involved in synaptic disorder in the absence of visible amyloid structures. In this study, the role of the human APP gene in synaptogenesis in transgenic lines of Drosophila melanogaster whose nerve cells express the human APP695 isoform, truncated APPs, and the presynaptic marker synaptotagmin driving the sequence of the green fluorescent protein. The expression of APP and its truncated forms caused a decrease in the synaptotagmin content of antennal lobes and mushroom lobes of the D. melanogaster brain, as well as neurodegeneration that progressed with age. The results suggest that that abnormal synaptogenesis and neurodegeneration occur in the Drosophila brain in the absence of Abeta. It is assumed that impaired cellular functions of APP and secretion of Abeta independently contribute to the pathogenesis of AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/biosynthesis , Drosophila melanogaster/metabolism , Synaptotagmins/metabolism , Alzheimer Disease/etiology , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Animals, Genetically Modified , Brain/metabolism , Brain/ultrastructure , Drosophila melanogaster/genetics , Drosophila melanogaster/ultrastructure , Green Fluorescent Proteins/genetics , Humans , Synapses/physiology , Synaptotagmins/genetics
18.
Anaesth Intensive Care ; 37(1): 38-45, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19157344

ABSTRACT

In this study, we assessed whether apolipoprotein E (APOE) polymorphism affects inflammatory responses and mortality in the caecal ligation and puncture model of peritonitis. In addition, we determined the effects of APOE mimetic peptide administration in this sepsis model. Differences in survival between targeted replacement mice expressing the human APOE3 allele (APOE3TR) and the APOE4 allele (APOE4TR) mice were assessed. In a separate series of experiments, COG1410, an apoE-mimetic peptide, was administered intravenously at 12-hour intervals for 72 hours and compared to vehicle-treated control animals. End-points included mortality and serum levels of interleukin-1beta, interleukin-6, interleukin-12 and tumour necrosis factor-alpha. Mice expressing the human APOE4 allele (n = 16) demonstrated an increase in mortality following caecal ligation and puncture compared with APOE3TR mice (n = 22; P = 0.039). Administration of the apolipoprotein E mimetic COG1410 was well tolerated and APOE3TR mice treated with peptide (n = 20) demonstrated a significant reduction in mortality compared with vehicle treated animals (n = 20; P = 0.007). A similar effect was also observed in APOE4TR animals, in which treatment with COG1410 was associated with reduced mortality compared with vehicle treatment (n =16 animals/group; P = 0.027). COG1410 was also associated with a reduction in TNFalpha, interleukin-1beta, interleukin-6 and interleukin-12 levels in both APOE3TR and APOE4TR (n = 5 animals/group) assessed at 24 hours. Thus, administration of an apolipoprotein E-mimetic peptide is well tolerated, suppresses inflammatory responses, and improves mortality in a caecal ligation and puncture model of sepsis.


Subject(s)
Apolipoprotein E3/genetics , Apolipoprotein E4/genetics , Apolipoproteins E/administration & dosage , Polymorphism, Genetic , Sepsis/genetics , Animals , Apolipoprotein E3/metabolism , Apolipoprotein E4/metabolism , Apolipoproteins E/therapeutic use , Disease Models, Animal , Genotype , Granulocyte-Macrophage Colony-Stimulating Factor/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interleukins/metabolism , Ligation , Mice , Mice, Inbred C57BL , Polymorphism, Genetic/genetics , Sepsis/drug therapy , Sepsis/mortality , Survival Rate , Treatment Outcome , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
19.
Euro Surveill ; 13(45): pii: 19032, 2008 Nov 06.
Article in English | MEDLINE | ID: mdl-19000568

ABSTRACT

The objective of this study was to estimate the lifetime age-specific cumulative incidence of self-reported genital warts diagnosis in Slovenia and to explore the association with demographic characteristics and self-reported sexual behaviour. Data were collected in the period from November 1999 to February 2001 from a national probability sample of the general population aged 18-49 years through a combination of face-to-face interviews at the respondents' homes and anonymous self-completed questionnaires. In total, 849 men and 903 women were interviewed (response: 63.3% men, 70.9% women). Among sexually experienced respondents with available information (752 men and 842 women), previous diagnosis of genital warts was reported by 0.3% of men (95% confidence interval (CI): 0.0%-1.3%) and 0.4% of women (95% CI: 0.1%-1.1%), and in the age group of 40-49 year-olds by 0.5% of men (95% CI:0.0-3.2) and 0.7% of women (95% CI: 0.2%-2.9%). In comparison to women with fewer than 10 lifetime male partners, those who reported to have had at least 10 male partners were more likely to have a previous diagnosis of genital warts (adjusted odds ratio: 7.2 (95% CI: 1.1%-47.8%). The lifetime cumulative incidence of self-reported genital warts diagnosis among Slovenians was relatively low in comparison to other published estimates from probability sample surveys in the general population in European countries. Our findings will inform the Slovenian vaccination policy against human papillomaviruses (HPV) and contribute to a better understanding of the differences between European countries regarding the burden of genital warts.


Subject(s)
Condylomata Acuminata/epidemiology , Data Collection , Data Interpretation, Statistical , Disease Outbreaks/statistics & numerical data , Population Surveillance , Risk Assessment/methods , Humans , Incidence , Risk Factors , Slovenia/epidemiology
20.
Biofizika ; 53(6): 1008-13, 2008.
Article in Russian | MEDLINE | ID: mdl-19137685

ABSTRACT

A comparative study of growth cone morphology in cultured embryonic neurons derived from wild type PS 1(+/+) and knockout PS 1(-/-) mice has been performed. Growth cones from wild type PS 1(+/+) mice were well spread and usually formed radially continuous and regular lamellar extensions with numerous filopodia. In contrast, most growth cones from knockout PS 1(-/-) mice exhibited small lamellar extensions, short filopodia, and pure adhesion. A significant amount of growth cones from knockout PS 1(-/-) mice collapsed after 3-4 days in culture. It was suggested that PS 1 plays an important role in growth cone structure by stabilizing the integrity of the cytoskeleton. The growth cone collapse may be the main reason for abnormal neuronal migration and impaired synaptic function in PS 1(-/-) mice.


Subject(s)
Growth Cones/ultrastructure , Neurons/ultrastructure , Presenilin-1/genetics , Animals , Cells, Cultured , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...