Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Fam Theory Rev ; 15(1): 57-77, 2023 Mar.
Article in English | MEDLINE | ID: mdl-38504801

ABSTRACT

Over the past two decades, scholars have investigated a multitude of different aspects of motherhood. This article provides a scoping review of research published from 2001 to 2021, covering 115 Social Science Citation Index-referenced papers from WEIRD countries, with the aim of reconstructing social norms around motherhood and mothers' responses to them. The analysis is theoretically based on normological and praxeological concepts. The findings reveal five contemporary norms of motherhood that reflect both stability and increasing differentiation, and are related to five types of mothers: the norms of being attentive to the child (present mother), of securing the child's successful development (future-oriented mother), of integrating employment into mothering (working mother), of being in control (public mother), and of being contented (happy mother). Relying on an intersectional lens, we analyze mothers' heterogeneous responses to these norms of motherhood, and examine how neoliberal demands build on and perpetuate inequalities.

2.
J Psychopharmacol ; 31(2): 204-210, 2017 02.
Article in English | MEDLINE | ID: mdl-27436232

ABSTRACT

Pro-inflammatory cytokines can promote sleep and neuronal processes underlying memory formation. However, this has mainly been revealed in animal studies. In this double-blind, placebo-controlled within-subject designed study, we examined how changes in the balance between pro- and anti-inflammatory signalling affect sleep and sleep-associated memory consolidation in humans. After learning declarative memory tasks (word pairs, texts) and a procedural memory task (finger tapping) in the evening, 21 healthy young men orally received either 200 mg of the anti-inflammatory antibiotic minocycline or placebo shortly before nocturnal sleep. Sleep was allowed between 23:00 and 07:00 h and recorded polysomnographically. Retrieval of memories was tested two days later. Because of outliers or missing data, final sample size was reduced to n = 14-19. Our data suggest that rather than weakening sleep as expected based on animal studies, the anti-inflammatory agent promoted sleep and memory consolidation. Specifically, minocycline increased slow-wave activity (0.68-4.0 Hz) during non-rapid eye movement sleep stage 2 and selectively enhanced episodic aspects in memory (i.e. memory for the temporal order of events in the texts). In combination with previous results, our findings indicate that, in humans, reducing pro-inflammatory signalling can act towards deepening non-rapid eye movement sleep and enhancing its memory forming efficacy.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Memory/drug effects , Minocycline/therapeutic use , Sleep/drug effects , Adult , Double-Blind Method , Humans , Learning/drug effects , Male , Memory Consolidation/drug effects , Young Adult
3.
Oncotarget ; 7(17): 24415-28, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-27027435

ABSTRACT

Sodium potassium pump (Na+/K+ ATPase) is a validated pharmacological target for the treatment of various cardiac conditions. Recent published data with Na+/K+ ATPase inhibitors suggest a potent anti-cancer action of these agents in multiple indications. In the present study, we focus on istaroxime, a Na+/K+ ATPase inhibitor that has shown favorable safety and efficacy properties in cardiac phase II clinical trials. Our experiments in 22 cancer cell lines and in prostate tumors in vivo proved the strong anti-cancer action of this compound. Istaroxime induced apoptosis, affected the key proliferative and apoptotic mediators c-Myc and caspase-3 and modified actin cystoskeleton dynamics and RhoA activity in prostate cancer cells. Interestingly, istaroxime was capable of binding to mAR, a membrane receptor mediating rapid, non-genomic actions of steroids in prostate and other cells. These results support a multi-level action of Na+/K+ ATPase inhibitors in cancer cells and collectively validate istaroxime as a strong re-purposing candidate for further cancer drug development.


Subject(s)
Etiocholanolone/analogs & derivatives , Prostatic Neoplasms/drug therapy , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Xenograft Model Antitumor Assays , Animals , Apoptosis/drug effects , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Clinical Trials, Phase II as Topic , Etiocholanolone/metabolism , Etiocholanolone/pharmacology , Female , HCT116 Cells , Humans , MCF-7 Cells , Male , Mice, Inbred NOD , Mice, SCID , Prostatic Neoplasms/metabolism , Protein Binding , Receptors, Androgen/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Tumor Burden/drug effects
4.
Arterioscler Thromb Vasc Biol ; 35(3): 655-63, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25550208

ABSTRACT

OBJECTIVE: Cyclophilin A (CyPA) is secreted under inflammatory conditions by various cell types. Whereas the important role of intracellular CyPA for platelet function has been reported, the effect of extracellular CyPA on platelet function has not been investigated yet. APPROACH AND RESULTS: Inhibition of extracellular CyPA through a novel specific inhibitor MM284 reduced thrombus after ferric chloride-induced injury in vivo. In vitro extracellular CyPA enhanced thrombus formation even in CyPA(-/-) platelets. Treatment of isolated platelets with recombinant CyPA resulted in platelet degranulation in a time- and dose-dependent manner. Inhibition of the platelet surface receptor extracellular matrix metalloproteinase inducer (cluster of differentiation 147) by an anticluster of differentiation 147 monoclonal antibody significantly reduced CyPA-dependent platelet degranulation. Pretreatment of platelets with CyPA enhanced their recruitment to mouse carotid arteries after arterial injury, which could be inhibited by an anticluster of differentiation 147 monoclonal antibody (intravital microscopy). The role of extracellular CyPA in adhesion could be confirmed by infusing CyPA(-/-) platelets in CyPA(+/+) mice and by infusing CyPA(+/+) platelets in CyPA(-/-) mice. Stimulation of platelets with CyPA induced phosphorylation of Akt, which could in turn be inhibited in the presence of phosphoinositid-3-kinase inhibitors. Akt-1(-/-) platelets revealed a markedly decreased degranulation on CyPA stimulation. Finally, ADP-induced platelet aggregation was attenuated by MM284, as well as by inhibiting paracrine-secreted CyPA without directly affecting Ca(2+)-signaling. CONCLUSIONS: Extracellular CyPA activates platelets via cluster of differentiation 147-mediated phosphoinositid-3-kinase/Akt-signaling, leading to enhanced adhesion and thrombus formation independently of intracellular CyPA. Targeting extracellular CyPA via a specific inhibitor may be a promising strategy for platelet inhibition without affecting critical functions of intracellular CyPA.


Subject(s)
Basigin/blood , Blood Platelets/enzymology , Cyclophilin A/blood , Phosphatidylinositol 3-Kinases/blood , Platelet Adhesiveness , Proto-Oncogene Proteins c-akt/blood , Signal Transduction , Thrombosis/enzymology , Animals , Blood Platelets/drug effects , Carotid Artery Injuries/blood , Carotid Artery Injuries/enzymology , Carotid Artery Injuries/genetics , Cell Degranulation/drug effects , Chlorides , Cyclophilin A/antagonists & inhibitors , Cyclophilin A/genetics , Disease Models, Animal , Dose-Response Relationship, Drug , Ferric Compounds , Fibrinolytic Agents/pharmacology , Humans , Mice, Inbred C57BL , Mice, Knockout , Platelet Adhesiveness/drug effects , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/deficiency , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/drug effects , Thrombosis/blood , Thrombosis/chemically induced , Thrombosis/genetics , Thrombosis/prevention & control , Time Factors
5.
Brain Behav Immun ; 47: 178-85, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25535859

ABSTRACT

Pro-inflammatory cytokines like interleukin-1 beta (IL-1) are major players in the interaction between the immune system and the central nervous system. Various animal studies report a sleep-promoting effect of IL-1 leading to enhanced slow wave sleep (SWS). Moreover, this cytokine was shown to affect hippocampus-dependent memory. However, the role of IL-1 in human sleep and memory is not yet understood. We administered the synthetic IL-1 receptor antagonist anakinra (IL-1ra) in healthy humans (100mg, subcutaneously, before sleep; n=16) to investigate the role of IL-1 signaling in sleep regulation and sleep-dependent declarative memory consolidation. Inasmuch monocytes have been considered a model for central nervous microglia, we monitored cytokine production in classical and non-classical blood monocytes to gain clues about how central nervous effects of IL-1ra are conveyed. Contrary to our expectation, IL-1ra increased EEG slow wave activity during SWS and non-rapid eye movement (NonREM) sleep, indicating a deepening of sleep, while sleep-associated memory consolidation remained unchanged. Moreover, IL-1ra slightly increased prolactin and reduced cortisol levels during sleep. Production of IL-1 by classical monocytes was diminished after IL-1ra. The discrepancy to findings in animal studies might reflect species differences and underlines the importance of studying cytokine effects in humans.


Subject(s)
Interleukin 1 Receptor Antagonist Protein/pharmacology , Memory Consolidation/drug effects , Monocytes/drug effects , Sleep/drug effects , Adolescent , Adrenocorticotropic Hormone/blood , Adult , Human Growth Hormone/blood , Humans , Hydrocortisone/blood , Male , Memory Consolidation/physiology , Prolactin/blood , Sleep/physiology , Young Adult
6.
FASEB J ; 28(5): 2108-19, 2014 May.
Article in English | MEDLINE | ID: mdl-24522202

ABSTRACT

Platelets are activated by increased cytosolic Ca(2+) concentration ([Ca(2+)]i) following store-operated calcium entry (SOCE) accomplished by calcium-release-activated calcium (CRAC) channel moiety Orai1 and its regulator STIM1. In other cells, Ca(2+) transport is regulated by 1,25(OH)2 vitamin D3 [1,25(OH)2D3]. 1,25(OH)2D3 formation is inhibited by klotho and excessive in klotho-deficient mice (kl/kl). The present study explored the effect of klotho deficiency on platelet Ca(2+) signaling and activation. Platelets and megakaryocytes isolated from WT and kl/kl-mice were analyzed by RT-PCR, Western blotting, confocal microscopy, Fura-2-fluorescence, patch clamp, flow cytometry, aggregometry, and flow chamber. STIM1/Orai1 transcript and protein levels, SOCE, agonist-induced [Ca(2+)]i increase, activation-dependent degranulation, integrin αIIbß3 activation and aggregation, and thrombus formation were significantly blunted in kl/kl platelets (by 27-90%). STIM1/Orai1 transcript and protein levels, as well as CRAC currents, were significantly reduced in kl/kl megakaryocytes (by 38-73%) and 1,25(OH)2D3-treated WT megakaryocytes. Nuclear NF-κB subunit p50/p65 abundance was significantly reduced in kl/kl-megakaryocytes (by 51-76%). Transfection with p50/p65 significantly increased STIM1/Orai1 transcript and protein levels in megakaryocytic MEG-01 cells (by 46-97%). Low-vitamin D diet (LVD) of kl/kl mice normalized plasma 1,25(OH)2D3 concentration and function of platelets and megakaryocytes. Klotho deficiency inhibits platelet Ca(2+) signaling and activation, an effect at least partially due to 1,25(OH)2D3-dependent down-regulation of NF-κB activity and STIM1/Orai1 expression in megakaryocytes.


Subject(s)
Blood Platelets/metabolism , Calcitriol/metabolism , Calcium Signaling , Calcium/metabolism , Glucuronidase/genetics , Thrombosis/metabolism , Animals , Calcium Channels/metabolism , Cell Nucleus/metabolism , Cytosol/metabolism , Down-Regulation , Klotho Proteins , Megakaryocytes/cytology , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , NF-kappa B/metabolism , ORAI1 Protein , Patch-Clamp Techniques , Platelet Aggregation , Signal Transduction , Stromal Interaction Molecule 1 , Transfection
7.
Arterioscler Thromb Vasc Biol ; 34(1): 61-71, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24233488

ABSTRACT

OBJECTIVE: Platelet activation is essential for primary hemostasis and acute thrombotic vascular occlusions. On activation, platelets release their prothrombotic granules and expose phosphatidylserine, thus fostering thrombin generation and thrombus formation. In other cell types, both degranulation and phosphatidylserine exposure are modified by sphingomyelinase-dependent formation of ceramide. The present study thus explored whether acid sphingomyelinase participates in the regulation of platelet secretion, phosphatidylserine exposure, and thrombus formation. APPROACH AND RESULTS: Collagen-related peptide-induced or thrombin-induced ATP release and P-selectin exposure were significantly blunted in platelets from Asm-deficient mice (Smpd1(-/-)) when compared with platelets from wild-type mice (Smpd1(+/+)). Moreover, phosphatidylserine exposure and thrombin generation were significantly less pronounced in Smpd1(-/-) platelets than in Smpd1(+/+) platelets. In contrast, platelet integrin αIIbß3 activation and aggregation, as well as activation-dependent Ca(2+) flux, were not significantly different between Smpd1(-/-) and Smpd1(+/+) platelets. In vitro thrombus formation at shear rates of 1700 s(-1) and in vivo thrombus formation after FeCl3 injury were significantly blunted in Smpd1(-/-) mice while bleeding time was unaffected. Asm-deficient platelets showed significantly reduced activation-dependent ceramide formation, whereas exogenous ceramide rescued diminished platelet secretion and thrombus formation caused by Asm deficiency. Treatment of Smpd1(+/+) platelets with bacterial sphingomyelinase (0.01 U/mL) increased, whereas treatment with functional acid sphingomyelinase-inhibitors, amitriptyline or fluoxetine (5 µmol/L), blunted activation-dependent platelet degranulation, phosphatidylserine exposure, and thrombus formation. Impaired degranulation and thrombus formation of Smpd1(-/-) platelets were again overcome by exogenous bacterial sphingomyelinase. CONCLUSIONS: Acid sphingomyelinase is a completely novel element in the regulation of platelet plasma membrane properties, secretion, and thrombus formation.


Subject(s)
Blood Platelets/enzymology , Cell Degranulation , Cell Membrane/enzymology , Platelet Activation , Sphingomyelin Phosphodiesterase/blood , Thrombosis/enzymology , Adenosine Triphosphate/blood , Animals , Blood Platelets/drug effects , Calcium/blood , Cell Degranulation/drug effects , Cell Membrane/drug effects , Ceramides/blood , Chlorides , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Ferric Compounds , Fibrinolytic Agents/pharmacology , Male , Mice , Mice, Knockout , P-Selectin/blood , Phosphatidylserines/blood , Platelet Activation/drug effects , Platelet Aggregation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/genetics , Thrombin/metabolism , Thrombosis/blood , Thrombosis/chemically induced , Thrombosis/genetics , Thrombosis/prevention & control , Time Factors
8.
J Geriatr Psychiatry Neurol ; 26(4): 221-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23904354

ABSTRACT

BACKGROUND: It is well known that burden among caregivers of patients with frontotemporal dementia (FTD) is high. However, little is known about the specific problems, the factors that contribute to caregiver burden, and the needs of the FTD caregivers-particularly those needs that are accessible by external support strategies. OBJECTIVE: We developed a standardized questionnaire that addressed burdens, problems, and the actual needs of FTD caregivers. A total of 94 caregivers were interviewed. RESULTS: It appears that changes in the patients' behavior and in the interpersonal relations between caregivers and patients are associated with caregiver depression. The most important needs and requests of the caregivers included information and psychosocial support through educated staff, financial support as well as the education of medical staff about the disease. CONCLUSIONS: Support strategies should focus on information and psychosocial support. Given the low prevalence of FTD, internet- and telephone-based strategies appear suitable.


Subject(s)
Adaptation, Psychological , Caregivers/psychology , Cost of Illness , Frontotemporal Dementia/diagnosis , Quality of Life , Activities of Daily Living , Adult , Aged , Aged, 80 and over , Depression/psychology , Female , Frontotemporal Dementia/psychology , Health Services Needs and Demand , Humans , Interpersonal Relations , Male , Mental Disorders , Middle Aged , Psychiatric Status Rating Scales , Social Support , Socioeconomic Factors , Surveys and Questionnaires
9.
Endocrinology ; 154(7): 2446-56, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23696568

ABSTRACT

Tumor growth is fostered by inhibition of cell death, which involves the receptiveness of tumor to growth factors and hormones. We have recently shown that testosterone exerts proapoptotic effects in prostate and colon cancer cells through a membrane-initiated mechanism. In addition, we have recently reported that dehydroepiandrosterone (DHEA) can control cell fate, activating nerve growth factor (NGF) receptors, namely tropomyosin-related kinase (Trk)A and p75 neurotrophin receptor, in primary neurons and in PC12 tumoral cells. NGF was recently involved in cancer cell proliferation and apoptosis. In the present study, we explored the cross talk between androgens (testosterone and DHEA) and NGF in regulating apoptosis of prostate and colon cancer cells. DHEA and NGF strongly blunted serum deprivation-induced apoptosis, whereas testosterone induced apoptosis of both cancer cell lines. The antiapoptotic effect of both DHEA and NGF was completely reversed by testosterone. In line with this, DHEA or NGF up-regulated, whereas testosterone down-regulated, the expression of TrkA receptor. The effects of androgens were abolished in both cell lines in the presence of TrkA inhibitor. DHEA induced the phosphorylation of TrkA and the interaction of p75 neurotrophin receptor with its effectors, Rho protein GDP dissociation inhibitor and receptor interacting serine/threonine-protein kinase 2. Conversely, testosterone was unable to activate both receptors. Testosterone acted as a DHEA and NGF antagonist, by blocking the activation of both receptors by DHEA or NGF. Our findings suggest that androgens may influence hormone-sensitive tumor cells via their cross talk with NGF receptors. The interplay between steroid hormone and neurotrophins signaling in hormone-dependent tumors offers new insights in the pathophysiology of these neoplasias.


Subject(s)
Dehydroepiandrosterone/pharmacology , Receptors, Nerve Growth Factor/metabolism , Testosterone/pharmacology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/metabolism , Humans , Male , Nerve Growth Factor/pharmacology , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , PC12 Cells , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Rats , Receptor, Nerve Growth Factor/genetics , Receptor, Nerve Growth Factor/metabolism , Receptors, Nerve Growth Factor/genetics , Signal Transduction/drug effects , Tumor Cells, Cultured
10.
Biochem Biophys Res Commun ; 436(1): 25-30, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23685155

ABSTRACT

Thrombin activates pore forming channel protein Orai1 resulting in store operated Ca(2+) entry (SOCE) with subsequent Ca(2+)-dependent release of platelet granules, activation of integrin αIIbß3, adhesion, aggregation and thrombus formation. Platelets lack nuclei and are thus unable to modify protein abundance by transcriptional regulation. Nevertheless, they still contain pre-mRNA and mRNA and are thus able to express protein by stimulation of rapid translation. Platelet translation is sensitive to phosphoinositide-3-kinase (PI3K) and actin polymerization. The present study explored whether platelet activation via thrombin modifies Orai1 protein abundance. According to RT-PCR platelets contain pre-mRNA and mRNA encoding Orai1. Activation with thrombin (0.1 U/ml) results in a significant decline of pre-mRNA, which is, according to Western blotting and confocal microscopy, paralleled by a marked and statistically significant increase of Orai1 protein abundance. The increase of Orai1 protein abundance is insensitive to inhibition of transcription with actinomycin (4 µg/ml), but is significantly blunted by inhibition of translation with puromycin (100 nM) and by inhibition of PI3K with wortmannin (100 nM) or LY294002 (25 µM). In conclusion, activation of platelets stimulates the translational expression of Orai1, thus augmenting platelet Ca(2+) signaling.


Subject(s)
Calcium Channels/metabolism , Gene Expression Regulation , Thrombin/metabolism , Androstadienes/pharmacology , Blood Platelets/metabolism , Calcium/metabolism , Chromones/pharmacology , Dactinomycin/pharmacology , Humans , Morpholines/pharmacology , ORAI1 Protein , Phosphatidylinositol 3-Kinases/metabolism , Platelet Activation , Puromycin/pharmacology , RNA Precursors/metabolism , RNA, Messenger/metabolism , Thrombosis/metabolism , Time Factors , Wortmannin
11.
FASEB J ; 27(7): 2799-806, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23568775

ABSTRACT

Chorea-acanthocytosis (ChAc), a lethal disease caused by defective chorein, is characterized by neurodegeneration and erythrocyte acanthocytosis. The functional significance of chorein in other cell types remained ill-defined. The present study revealed chorein expression in blood platelets. As compared to platelets from healthy volunteers, platelets from patients with ChAc displayed a 47% increased globular/filamentous actin ratio, indicating actin depolymerization. Moreover, phosphoinositide-3-kinase subunit p85 phosphorylation, p21 protein-activated kinase (PAK1) phosphorylation, as well as vesicle-associated membrane protein 8 (VAMP8) expression were significantly reduced in platelets from patients with ChAc (by 17, 22, and 39%, respectively) and in megakaryocytic (MEG-01) cells following chorein silencing (by 16, 54, and 11%, respectively). Activation-induced platelet secretion from dense granules (ATP release) and α granules (P-selectin exposure) were significantly less (by 55% after stimulation with 1 µg/ml CRP and by 33% after stimulation with 5 µM TRAP, respectively) in ChAc platelets than in control platelets. Furthermore, platelet aggregation following stimulation with different platelet agonists was significantly impaired. These observations reveal a completely novel function of chorein, i.e., regulation of secretion and aggregation of blood platelets.


Subject(s)
Blood Platelets/metabolism , Cell Degranulation , Cytoskeleton/metabolism , Vesicular Transport Proteins/metabolism , Actins/metabolism , Adult , Blood Platelets/physiology , Blood Platelets/ultrastructure , Blotting, Western , Cell Line, Tumor , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Female , Humans , Male , Microscopy, Confocal , Microscopy, Electron, Transmission , Middle Aged , Neuroacanthocytosis/blood , Neuroacanthocytosis/genetics , Neuroacanthocytosis/metabolism , Phosphorylation , Platelet Aggregation , R-SNARE Proteins/metabolism , RNA Interference , Vesicular Transport Proteins/genetics , Young Adult , p21-Activated Kinases/metabolism
12.
Biochem Biophys Res Commun ; 435(2): 323-6, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23399563

ABSTRACT

Glycopeptides, such as vancomycin, are powerful antibiotics against methicillin-resistant Staphylococcus aureus. Balhimycin, a glycopeptide antibiotic isolated from Amycolatopsis balhimycina, is similarly effective as vancomycin. Side effects of vancomycin include triggering of platelet apoptosis, which is characterized by cell shrinkage and by cell membrane scrambling with phosphatidylserine exposure at the cell surface. Stimulation of apoptosis may involve increase of cytosolic Ca(2+) activity, ceramide formation, mitochondrial depolarization and/or caspase activation. An effect of balhimycin on apoptosis has, however, never been reported. The present study thus tested whether balhimycin triggers platelet apoptosis. Human blood platelets were treated with balhimycin and cell volume was estimated from forward scatter, phosphatidylserine exposure from annexin V-binding, cytosolic Ca(2+) activity from fluo-3AM fluorescence, ceramide formation utilizing antibodies, mitochondrial potential from DiOC6 fluorescence, and caspase-3 activity utilizing antibodies. As a result, a 30 min exposure to balhimycin significantly decreased cell volume (≥1 µg/ml), triggered annexin V binding (≥1 µg/ml), increased cytosolic Ca(2+) activity (≥1 µg/ml), stimulated ceramide formation (≥10 µg/ml), depolarized mitochondria (≥1 µg/ml) and activated caspase-3 (≥1 µg/ml). Cell membrane scrambling and caspase-3 activation were virtually abrogated by removal of extracellular Ca(2+). Cell membrane scrambling was not significantly blunted by pancaspase inhibition with zVAD-FMK (1µM). In conclusion, balhimycin triggers cell membrane scrambling of platelets, an effect dependent on Ca(2+), but not on activation of caspases.


Subject(s)
Blood Platelets/physiology , Calcium/metabolism , Ceramides/metabolism , Membrane Fluidity/drug effects , Membrane Fluidity/physiology , Vancomycin/analogs & derivatives , Adult , Anti-Bacterial Agents/administration & dosage , Apoptosis/drug effects , Apoptosis/physiology , Blood Platelets/drug effects , Cell Size , Cells, Cultured , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Vancomycin/administration & dosage
13.
Cell Physiol Biochem ; 31(1): 102-12, 2013.
Article in English | MEDLINE | ID: mdl-23363637

ABSTRACT

BACKGROUND/AIMS: Side effects of vancomycin, a widely used antibiotic, include thrombocytopenia. The vancomycin-induced thrombocytopenia has been attributed to immune reactions. At least in theory, thrombocytopenia could result in part from the triggering of apoptosis, which results in cell shrinkage and cell membrane scrambling with subsequent phosphatidylserine exposure at the cell surface. The cell membrane scrambling could be initiated by a signaling involving increase of cytosolic Ca(2+) activity, ceramide formation, mitochondrial depolarization and/or caspase activation. Vancomycin has indeed been shown to trigger neutrophil apoptosis. An effect of vancomycin on platelet apoptosis has, however, never been tested. The present study thus explored the effect of vancomycin on platelet activation and apoptosis. METHODS: Human blood platelets were exposed to vancomycin and forward scatter was utilized to estimate cell volume, annexin V-binding to quantify phosphatidylserine (PS) exposure, Fluo-3 AM fluorescence to estimate cytosolic Ca(2+) activity ([Ca(2+)]i), antibodies to quantify ceramide formation and immunofluorescence to quantify protein abundance of active caspase-3. RESULTS: A 30 minutes exposure to vancomycin (≥1 µg/ ml) decreased cell volume, triggered annexin V-binding, increased [Ca(2+)]i, activated caspase 3, stimulated ceramide formation, triggered release of thromboxane B2, and upregulated surface expression of CD62P (P-selectin) as well as activated integrin αllbß3. Annexin V-binding and upregulation of CD62P (P-selectin) and integrin αllbß3 was significantly blunted by removal of extracellular Ca(2+). Annexin V-binding was not significantly blunted by pan-caspase inhibitor zVAD-FMK (1 µM). In conclusion, vancomycin results in platelet activation and suicidal platelet death with increase of [Ca(2+)]i, caspase-3 activation, cell membrane scrambling and cell shrinkage. Activation and cell membrane scrambling required the presence of Ca(2+), but not activation of caspases. CONCLUSION: Vancomycin exposure leads to platelet activation and apoptosis.


Subject(s)
Anti-Bacterial Agents/pharmacology , Apoptosis/drug effects , Blood Platelets/drug effects , Vancomycin/pharmacology , Adult , Amino Acid Chloromethyl Ketones/pharmacology , Aniline Compounds/chemistry , Annexin A5/metabolism , Blood Platelets/cytology , Calcium/metabolism , Caspase 3/metabolism , Caspase Inhibitors/pharmacology , Cell Size/drug effects , Ceramides/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Middle Aged , P-Selectin/metabolism , Phosphatidylserines/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Binding , Thromboxane B2/metabolism , Xanthenes/chemistry
14.
Cell Signal ; 25(1): 66-73, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23316499

ABSTRACT

Actin cytoskeleton reorganization initiated by testosterone conjugates through activation of membrane androgen receptors (mAR) has recently been reported in colon tumor cells. This mAR-induced actin reorganization was recognized as a critical initial event, controlling apoptosis and inhibiting cell migration. The present study addressed the molecular signaling regulating the rapid actin remodeling initiated upon testosterone-induced mAR activation in Caco2 colon tumor cells. We report early phosphorylation of the Focal Adhesion Kinase (FAK), followed by substantial early phosphorylation of mammalian target of rapamycin (mTOR), S6 kinase (p70S6K) and the actin regulating p21-activated kinase (PAK1). Pharmacological inhibition of FAK-sensitive phosphatidylinositide-3-kinase (PI-3K), a known element of mAR-signaling, fully abrogated the testosterone-induced actin reorganization and the activation of mTOR, p70S6K and PAK1. Similarly, inhibition of mTOR blocked p70S6K and PAK1 phosphorylation and actin remodeling. Pretreatment of the cells with the intracellular androgen receptor (iAR) antagonist flutamide or silencing iAR through siRNA did not influence mTOR phosphorylation and actin reorganization, indicating specific mAR-induced testosterone effects that are independent of iAR signaling. In conclusion, we demonstrate for the first time a new mAR-governed pathway involving FAK/PI-3K and mTOR/p70S6K/PAK1-cascade that regulates early actin reorganization in colon cancer cells.


Subject(s)
Actin Cytoskeleton/drug effects , Actins/metabolism , Receptors, Androgen/metabolism , Signal Transduction , Testosterone/pharmacology , Androgen Receptor Antagonists/pharmacology , Caco-2 Cells , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Flutamide/pharmacology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Serum Albumin/chemistry , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Testosterone/chemistry , p21-Activated Kinases/metabolism , rho-Associated Kinases/metabolism
15.
Circ Res ; 111(10): 1297-307, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22927331

ABSTRACT

RATIONALE: The recently discovered chemokine CXC motif ligand 16 (CXCL16) is highly expressed in atherosclerotic lesions and is a potential pathogenic mediator in coronary artery disease. OBJECTIVE: The aim of this study was to test the role of CXCL16 on platelet activation and vascular adhesion, as well as the underlying mechanism and signaling pathway. METHODS AND RESULTS: Reverse-transcriptase polymerase chain reaction, Western blotting, confocal microscopy, and flow cytometry revealed that CXCL16-specific receptor, CXC motif receptor 6, is highly expressed in platelets. According to flow cytometry and confocal microscopy, stimulation of platelets with CXCL16 induced platelet degranulation, integrin α(IIb)ß(3) activation, and shape change. CXCL16 increased Akt phosphorylation (Thr(308)/Ser(473)), an effect abrogated by phosphatidylinositide 3-kinase inhibitors wortmannin (100 nmol/L) and LY294002 (25 µmol/L). The phosphatidylinositide 3-kinase inhibitors and Akt inhibitor SH-6 (20 µmol/L) further diminished CXCL16-induced platelet activation. CXCL16-mediated platelet degranulation, integrin α(IIb)ß(3) activation, and Akt phosphorylation were blunted in platelets lacking CXCL16-specific receptor CXC motif receptor 6. CXCL16-induced platelet activation was abrogated in Akt1- or Akt2-deficient platelets. CXCL16 enhanced platelet adhesion to endothelium in vitro after high arterial shear stress (2000(-s)) and to injured vascular wall in vivo after carotid ligation. CXCL16-induced stimulation of platelet adhesion again was prevented by phosphatidylinositide 3-kinase and Akt inhibitors. Apyrase and antagonists of platelet purinergic receptors P(2)Y(1) (MRS2179, 100 µmol/L) and especially P(2)Y(12) (Cangrelor, 10 µmol/L) blunted CXCL16-triggered platelet activation as well as CXCL16-induced platelet adhesion under high arterial shear stress in vitro and after carotid ligation in vivo. CONCLUSIONS: The inflammatory chemokine CXCL16 triggers platelet activation and adhesion via CXC motif receptor 6-dependent phosphatidylinositide 3-kinase/Akt signaling and paracrine activation, suggesting a decisive role for CXCL16 in linking vascular inflammation and thrombo-occlusive diseases.


Subject(s)
Chemokine CXCL6/immunology , Chemokine CXCL6/metabolism , Platelet Activation/immunology , Platelet Adhesiveness/immunology , Signal Transduction/immunology , Animals , Benzofurans , Blood Platelets/immunology , Blood Platelets/metabolism , Chemokine CXCL16 , Coronary Artery Disease/immunology , Coronary Artery Disease/metabolism , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Female , Humans , Male , Mice , Mice, Mutant Strains , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Quinolines , Receptors, CXCR/genetics , Receptors, CXCR/metabolism , Receptors, CXCR6 , Thrombosis/immunology , Thrombosis/metabolism , Vasculitis/immunology , Vasculitis/metabolism
16.
Biochem Biophys Res Commun ; 425(1): 1-5, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22809514

ABSTRACT

Activation of platelets by thrombin opens pore forming channel protein Orai1 with subsequent store operated Ca(2+) entry (SOCE) and Ca(2+) dependent platelet granule release, integrin α(IIb)ß(3) activation, adhesion, aggregation and thrombus formation. Orai1 and thus SOCE as well as platelet activation are up-regulated by the serum- and glucocorticoid-inducible kinase-1 (SGK1), which transcriptionally regulates Orai1 expression in megakaryocytes and thus determines Orai1 protein abundance in mature, circulating platelets. As platelets are devoid of nuclei, they are unable to modify protein abundance by regulation of transcription. However, they contain mRNA and thus could express novel protein by stimulation of protein translation. Translation is sensitive to actin polymerization and phosphoinositide-3-kinase (PI3K). Translational regulation of SGK1 expression has never been described before. The present study thus explored whether thrombin regulates SGK1 expression in platelets. As a result, according to RT-PCR mRNA encoding SGK1 is present in circulating platelets and significantly decreased by activation of platelets with thrombin (1 U/ml). The protein abundance of SGK1 is significantly enhanced by thrombin treatment, an effect significantly decreased by inhibition of translation with puromycin (100 nM) but not by inhibition of transcription with actinomycin (4 µg/ml). The increase of SGK1 protein abundance is blunted by inhibition of PI3K with wortmannin (100 nM) or LY294002 (25 µM), and by disruption of the cytoskeleton with cytochalasin B (1 µM). In conclusion, activation of platelets with thrombin stimulates the translation of SGK1.


Subject(s)
Blood Platelets/enzymology , Immediate-Early Proteins/biosynthesis , Platelet Activation/physiology , Protein Biosynthesis/physiology , Protein Serine-Threonine Kinases/biosynthesis , Thrombin/physiology , Androstadienes/pharmacology , Calcium Channels/biosynthesis , Cells, Cultured , Chromones/pharmacology , Cytochalasin B/pharmacology , Humans , Immediate-Early Proteins/genetics , Morpholines/pharmacology , ORAI1 Protein , Phosphodiesterase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Platelet Activation/drug effects , Platelet Activation/genetics , Protein Biosynthesis/drug effects , Protein Biosynthesis/genetics , Protein Serine-Threonine Kinases/genetics , Thrombin/pharmacology , Transcription, Genetic , Wortmannin
17.
Cell Physiol Biochem ; 30(1): 259-68, 2012.
Article in English | MEDLINE | ID: mdl-22759972

ABSTRACT

Recent observations pointed to the ability of platelets to migrate and thus to invade the inflamed vascular wall. Platelet migration could be stimulated by stromal cell-derived factor-1 (SDF-1), an effect dependent on phosphatidylinositide-3-kinase (PI3K) and paralleled by activation and phosphorylation of Wiskott-Aldrich syndrome protein (WASP). Migration is inhibited by vinculin, which is similarly regulated by phosphorylation. PI3K-sensitive kinases include the serum- and glucocorticoid-inducible kinase 1 (SGK1). The present study explored whether SGK1 modifies WASP and vinculin phosphorylation in murine platelets and participates in the regulation of platelet migration. Platelets were isolated from gene-targeted mice lacking SGK1 (sgk1(-/-)) and from their wild type littermates (sgk1(+/+)). Platelet migration stimulated with SDF-1 was significantly less pronounced in sgk1(-/-)platelets than in sgk1(+/+) platelets. Moreover, SDF-1 significantly induced WASP phosphorylation, an effect again reduced in platelets lacking SGK1. Phosphorylation of vinculin was significantly enhanced in sgk1(-/-)platelets and was significantly reduced following treatment of platelets with Ca(2+) chelator BAPTA. Immunohistochemical analysis of in vivo experiments in intestinal vessels after vascular inflammation revealed that transmigration of platelets into inflamed vessel walls was significantly less pronounced in sgk1(-/-)than in sgk1(+/+) mice. In conclusion, SGK1 is a powerful regulator of platelet migration.


Subject(s)
Blood Platelets/physiology , Immediate-Early Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Transendothelial and Transepithelial Migration , Animals , Cells, Cultured , Chelating Agents/pharmacology , Chemokine CXCL12/physiology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Female , Immediate-Early Proteins/genetics , Intestines/blood supply , Ischemia/metabolism , Ischemia/pathology , Male , Mice , Mice, Knockout , Phosphorylation , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Vinculin/metabolism , Wiskott-Aldrich Syndrome Protein/metabolism
18.
Apoptosis ; 17(9): 998-1008, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22752708

ABSTRACT

Peptidoglycan (PGN), a component of bacterial cell wall and belonging to "Microbe-Associated Molecular Patterns" (MAMP) triggers host reactions contributing to the pathophysiology of infectious disease. Host cell responses to PGN exposure include apoptosis. Bacterial infections may result in activation of blood platelets and thrombocytopenia. The present study explored, whether HPLC-purified fractions of PGNs from Staphylococcus aureus 113 triggers apoptosis of platelets. To this end platelets were exposed to PGN fractions and annexin-V binding determined to depict cell membrane scrambling, DiOC6 fluorescence to estimate depolarization of mitochondrial potential, Fluo-3AM staining for intracellular Ca(2+) activity ([Ca(2+)](i)) and immunofluorescence to quantify protein abundance of active caspase-3. As a result, a 30 min exposure to monomeric fraction (mPGN) (≥50 ng/ml) was followed by annexin-V binding, paralleled by increase of [Ca(2+)](i), mitochondrial depolarization, caspase-3 activation and integrin α(IIb)ß(3) upregulation. The annexin-V binding was significantly blunted by anti-TLR-2 antibodies, in absence of extracellular Ca(2+), and by pancaspase inhibitor zVAD-FMK (1 µM). In conclusion, PGN triggers apoptosis of platelets in activation-dependent manner, characterized by mitochondrial depolarization, caspase-3 activation and cell membrane scrambling.


Subject(s)
Apoptosis/drug effects , Blood Platelets/drug effects , Blood Platelets/metabolism , Peptidoglycan/pharmacology , Staphylococcus aureus/chemistry , Amino Acid Chloromethyl Ketones/pharmacology , Annexin A5/metabolism , Antibodies/immunology , Caspase 3/metabolism , Caspase Inhibitors/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Enzyme Activation , Humans , Integrin alpha2/biosynthesis , Mitochondria/metabolism , Toll-Like Receptor 2/immunology
19.
J Mol Cell Cardiol ; 53(1): 6-14, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22446162

ABSTRACT

Extracellular cyclophilin A (CyPA) and its receptor Extracellular Matrix Metalloproteinase Inducer (EMMPRIN, CD147) modulate inflammatory processes beyond metalloproteinase (MMP) activity. Recently, we have shown that CyPA and CD147 are upregulated in patients with inflammatory cardiomyopathy. Here we investigate the role of CyPA and CD147 in murine coxsackievirus B3 (CVB3)-induced myocarditis. CVB3-infected CyPA(-/-) mice (129S6/SvEv) revealed a significantly reduced T-cell and macrophage recruitment at 8 days p.i. compared to wild-type mice. In A.BY/SnJ mice, treatment with the cyclophilin-inhibitor NIM811 was associated with a reduction of inflammatory lesions and MMP-9 expression but with enhanced virus replication 8 days p.i. At 28 days p.i. the extent of lesion areas was not affected bei NIM811, whereas the collagen content was reduced. Initiation of NIM811-treatment on day 12 (after an effective virus defense) resulted in an even more pronounced reduction of myocardial fibrosis. In conclusion, in CVB3-induced myocarditis CyPA is important for macrophage and T cell recruitment and effective virus defense and may represent a pharmacological target to modulate myocardial remodeling in myocarditis.


Subject(s)
Coxsackievirus Infections/immunology , Cyclophilin A/metabolism , Endomyocardial Fibrosis/immunology , Enterovirus B, Human/immunology , Myocarditis/immunology , Animals , Basigin/metabolism , Cell Line , Cyclophilin A/antagonists & inhibitors , Cyclophilin A/deficiency , Cyclosporine/pharmacology , Endomyocardial Fibrosis/etiology , Endomyocardial Fibrosis/pathology , Humans , Macrophages/drug effects , Macrophages/immunology , Matrix Metalloproteinase 9/metabolism , Mice , Mice, 129 Strain , Mice, Knockout , Monocytes/drug effects , Monocytes/immunology , Myocarditis/complications , Myocarditis/virology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Ventricular Remodeling/drug effects
20.
FEBS J ; 279(7): 1231-42, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22309306

ABSTRACT

The serum- and glucocorticoid-dependent kinases 1-3 (SGK1-3) are downstream effectors of phosphatidylinositol 3-kinases, implicated in various cell responses including colon cancer tumorigenesis in mice. Here, we investigated the role of SGK1 in the regulation of cell motility. Using Caco-2 colon tumor and HEK293 embryonic kidney cells, we report that transfection with the constitutively active SGK1 mutant (SGK1-SD) significantly enhanced cell motility. The cell-adhesion protein vinculin was effectively dephosphorylated in SGK1-SD-transfected cells. Treatment of the cells with phosphatase inhibitors restored vinculin phosphorylation and inhibited cell migration, indicating a significant role for vinculin phosphorylation in SGK1-induced motility. SGK1-SD-enhanced cell motility was inhibited by activation of membrane androgen-binding sites (mAR) via testosterone-conjugates in both cell lines, whereas intracellular androgen receptor (iAR)-silencing and flutamide treatment revealed that these effects were clearly independent of the interaction of SGK1 with the classical androgen receptors (iAR). More importantly, mAR activation restored vinculin phosphorylation in SGK1-SD-transfected cells, whereas silencing of vinculin fully reversed the mAR-induced inhibition of the migratory capacity, implying that this protein is directly involved in cell motility regulation by SGK1 and mAR. This study indicates for the first time that SGK1 regulates cell migration via vinculin dephosphorylation, a mechanism that is controlled by mAR function.


Subject(s)
Cell Movement/physiology , Immediate-Early Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Androgen/metabolism , Vinculin/metabolism , Animals , Caco-2 Cells , HEK293 Cells , Humans , Immediate-Early Proteins/genetics , Mice , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Receptors, Androgen/genetics , Vinculin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...