Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 2537, 2023 05 03.
Article in English | MEDLINE | ID: mdl-37137944

ABSTRACT

The genomes of most protozoa encode families of variant surface antigens. In some parasitic microorganisms, it has been demonstrated that mutually exclusive changes in the expression of these antigens allow parasites to evade the host's immune response. It is widely assumed that antigenic variation in protozoan parasites is accomplished by the spontaneous appearance within the population of cells expressing antigenic variants that escape antibody-mediated cytotoxicity. Here we show, both in vitro and in animal infections, that antibodies to Variant-specific Surface Proteins (VSPs) of the intestinal parasite Giardia lamblia are not cytotoxic, inducing instead VSP clustering into liquid-ordered phase membrane microdomains that trigger a massive release of microvesicles carrying the original VSP and switch in expression to different VSPs by a calcium-dependent mechanism. This novel mechanism of surface antigen clearance throughout its release into microvesicles coupled to the stochastic induction of new phenotypic variants not only changes current paradigms of antigenic switching but also provides a new framework for understanding the course of protozoan infections as a host/parasite adaptive process.


Subject(s)
Giardia lamblia , Giardiasis , Intestinal Diseases, Parasitic , Parasites , Animals , Giardia lamblia/genetics , Giardia lamblia/metabolism , Parasites/metabolism , Antigens, Surface/genetics , Antigens, Surface/metabolism , Antigens, Protozoan , Antibodies/metabolism , Antigenic Variation , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
2.
Food Chem Toxicol ; 158: 112649, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34728246

ABSTRACT

Phthalates are synthetic chemicals widely used to make polyvinylchloride (PVC) soft and flexible. Of these, Di-(2-ethylhexyl) phthalate (DEHP) is the most commonly used, with high human exposure occurring as early as the fetal developmental stage and affecting the endocrine system. We focused on the perinatal DEHP effects on pituitary estrogen receptor (ER) expression in male rats, explored their impact on lactotroph and somatotroph cell growth, and evaluated the direct effects of this phthalate on pituitary cell cultures. Our results showed that DEHP perinatal exposure was unable to modify the ERα+ pituitary cell number from prepuberal rats, but increased ERß+ cells. In adulthood, the pituitary ERα+ cells underwent a slight decrease with ERß showing the greatest changes, and with a significant increase observed in somatotroph cells. Also, in vitro, DEHP reduced the ERα+ cells, increased the percentage of ERß+ pituitary cells and modified the Ki67 index, as well as decreasing the lactotrophs and increasing the somatotroph cells. In conclusion, the present study showed that DEHP induced ER expression changes in normal pituitary glands from male rats in in vivo and in vitro conditions, suggesting that DEHP could differentially modulate lactotroph and somatotroph cell growth, possibly as a consequence of ER imbalance.


Subject(s)
Diethylhexyl Phthalate/toxicity , Endocrine Disruptors/toxicity , Pituitary Gland , Prenatal Exposure Delayed Effects , Receptors, Estrogen/metabolism , Animals , Cell Proliferation/drug effects , Female , Lactotrophs/drug effects , Lactotrophs/metabolism , Male , Pituitary Gland/cytology , Pituitary Gland/drug effects , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/metabolism , Rats , Rats, Wistar , Somatotrophs/drug effects , Somatotrophs/metabolism
3.
J Cell Physiol ; 236(10): 6974-6987, 2021 10.
Article in English | MEDLINE | ID: mdl-33682941

ABSTRACT

Octreotide (OCT) is used to inhibit hormone secretion and growth in somatotroph tumors, although a significant percentage of patients are resistant. It has also been tested in nonfunctioning (NF) tumors but with poor results, with these outcomes having been associated with SSTR2 levels and impaired signaling. We investigated whether OCT inhibitory effects can be improved by TGF-ß1 in functioning and nonfunctioning somatotroph tumor cells. OCT effects on hormone secretion and proliferation were analyzed in the presence of TGF-ß1 in WT and SSTR2-overexpressing secreting GH3 and silent somatotroph tumor cells. The mechanism underlying these effects was assessed by studying SSTR and TGFßR signaling pathways mediators. In addition, we analyzed the effects of OCT/TGF-ß1 treatment on tumor growth and cell proliferation in vivo. The inhibitory effects of OCT on GH- and PRL-secretion and proliferation were improved in the presence of TGF-ß1, as well as by SSTR2 overexpression. The OCT/TGF-ß1 treatment induced downregulation of pERK1/2 and pAkt, upregulation of pSmad3, and inhibition of cyclin D1. In vivo experiments showed that OCT in the presence of TGF-ß1 blocked tumor volume growth, decreased cell proliferation, and increased tumor necrosis. These results indicate that SSTR2 levels and the stimulation of TGF-ß1/TGFßR/Smad2/3 pathway are important for strengthening the antiproliferative and antisecretory effects of OCT.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Cell Proliferation/drug effects , Octreotide/pharmacology , Pituitary Neoplasms/drug therapy , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Somatotrophs/drug effects , Transforming Growth Factor beta1/pharmacology , Animals , Cell Line , Female , Humans , Mice, Nude , Phosphorylation , Pituitary Neoplasms/genetics , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Rats , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Signal Transduction , Somatotrophs/metabolism , Somatotrophs/pathology , Tumor Burden/drug effects
4.
Nat Commun ; 10(1): 361, 2019 01 21.
Article in English | MEDLINE | ID: mdl-30664644

ABSTRACT

Intestinal and free-living protozoa, such as Giardia lamblia, express a dense coat of variant-specific surface proteins (VSPs) on trophozoites that protects the parasite inside the host's intestine. Here we show that VSPs not only are resistant to proteolytic digestion and extreme pH and temperatures but also stimulate host innate immune responses in a TLR-4 dependent manner. We show that these properties can be exploited to both protect and adjuvant vaccine antigens for oral administration. Chimeric Virus-like Particles (VLPs) decorated with VSPs and expressing model surface antigens, such as influenza virus hemagglutinin (HA) and neuraminidase (NA), are protected from degradation and activate antigen presenting cells in vitro. Orally administered VSP-pseudotyped VLPs, but not plain VLPs, generate robust immune responses that protect mice from influenza infection and HA-expressing tumors. This versatile vaccine platform has the attributes to meet the ultimate challenge of generating safe, stable and efficient oral vaccines.


Subject(s)
Giardia lamblia/chemistry , Influenza Vaccines/immunology , Membrane Proteins/immunology , Orthomyxoviridae Infections/prevention & control , Protozoan Proteins/immunology , Vaccines, Virus-Like Particle/immunology , Adjuvants, Immunologic , Administration, Oral , Animals , Antigen Presentation/drug effects , Bioengineering/methods , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/virology , Female , Gene Expression , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Immunity, Innate/drug effects , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Male , Membrane Proteins/genetics , Mice , Mice, Transgenic , Neuraminidase/genetics , Neuraminidase/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Protein Stability , Protozoan Proteins/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Trophozoites/chemistry , Vaccination , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/genetics
5.
J Endocrinol ; 240(2): 229-241, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30400032

ABSTRACT

The molecular mechanisms underlying the ERα nuclear/cytoplasmic pool that modulates pituitary cell proliferation have been widely described, but it is still not clear how ERα is targeted to the plasma membrane. The aim of this study was to analyse ERα palmitoylation and the plasma membrane ERα (mERα) pool, and their participation in E2-triggered membrane-initiated signalling in normal and pituitary tumour cell growth. Cell cultures were prepared from anterior pituitaries of female Wistar rats and tumour GH3 cells, and treated with 10 nM of oestradiol (E2). The basal expression of ERα was higher in tumour GH3 than in normal pituitary cells. Full-length palmitoylated ERα was observed in normal and pituitary tumour cells, demonstrating that E2 stimulation increased both, ERα in plasma membrane and ERα and caveolin-1 interaction after short-term treatment. In addition, the Dhhc7 and Dhhc21 palmitoylases were negatively regulated after sustained stimulation of E2 for 3 h. Although the uptake of BrdU into the nucleus in normal pituitary cells was not modified by E2, a significant increase in the GH3 tumoural cell, as well as ERK1/2 activation, with this effect being mimicked by PPT, a selective antagonist of ERα. These proliferative effects were blocked by ICI 182780 and the global inhibitor of palmitoylation. These findings indicate that ERα palmitoylation modulated the mERα pool and consequently the ERK1/2 pathway, thereby contributing to pituitary tumour cell proliferation. These results suggest that the plasma membrane ERα pool might be related to the proliferative behaviour of prolactinoma and may be a marker of pituitary tumour growth.


Subject(s)
Cell Membrane/metabolism , Cell Proliferation , Estrogen Receptor alpha/metabolism , Pituitary Neoplasms/metabolism , Animals , Antineoplastic Agents, Hormonal/pharmacology , Cell Line, Tumor , Cell Membrane/drug effects , Cells, Cultured , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogens/pharmacology , Female , Fulvestrant/pharmacology , Gene Expression/drug effects , Lipoylation/drug effects , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Rats, Wistar
6.
J Cell Physiol ; 233(2): 1402-1413, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28542730

ABSTRACT

In this study, we focused on ERß regulation in the adenohypophysis under different estrogenic milieu, by analyzing whether ER modulates the phosphatase and tensin homolog deleted on chromosome 10 (PTEN) expression and its subcellular localization on anterior pituitary glands from Wistar rats and GH3 lactosomatotroph cells that over-expressed ERß. ERß was regulated in a cyclic manner, and underwent dynamic changes throughout the estrous cycle, with decreased ERß+ cells in estrus and under E2 treatment, but increased in ovariectomized rats. In addition, the ERα/ß ratio increased in estrus and under E2 stimulation, but decreased in ovariectomized rats. Double immunofluorescence revealed that lactotroph and somatotroph ERß+ were significantly decreased in estrus. Also, variations in the PTEN expression was observed, which was diminished with high E2 conditions but augmented with low E2 milieu. The subcellular localization of this phosphatase was cell cycle-dependent, with remarkable changes in the immunostaining pattern: nuclear in arrested pituitary cells but cytoplasmic in stimulated cells, and responding differently to ER agonists, with only DPN being able to increase PTEN expression and retaining it in the nucleus. Finally, ERß over-expression increased PTEN with a noticeable subcellular redistribution, and with a significant nuclear signal increase in correlation with an increase of cells in G0/G1 phase. These results showed that E2 is able to inhibit ERß expression and suggests that the tumoral suppressor PTEN might be one of the signaling proteins by which E2, through ERß, acts to modulate pituitary cell proliferation, thereby adapting endocrine populations in relation with hormonal necessities.


Subject(s)
Cell Proliferation , Estrogen Receptor beta/metabolism , Estrous Cycle/metabolism , Lactotrophs/enzymology , PTEN Phosphohydrolase/metabolism , Somatotrophs/enzymology , Animals , Cells, Cultured , Estradiol/metabolism , Estradiol/pharmacology , Estrogen Receptor beta/agonists , Estrogen Receptor beta/genetics , Estrogen Replacement Therapy , Female , G1 Phase , Lactotrophs/drug effects , Male , Nitriles/pharmacology , Ovariectomy , Rats, Wistar , Signal Transduction , Somatotrophs/drug effects , Transfection
7.
Mol Cell Endocrinol ; 415: 100-13, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26282612

ABSTRACT

Considering that the role of ERß in the growth of pituitary cells is not well known, the aim of this work was to determine the expression of ERß in normal and tumoral cells and to investigate its implications in the proliferative control of this endocrine gland, by analyzing the participation of cyclin D1, Cdk4 and p21. Our results showed that the expression of ERß decreased during pituitary tumoral development induced by chronic E2 stimulation. The 20 ± 1.6% of normal adenohypophyseal cells expressed ERß, with this protein being reduced in the hyperplastic/adenomatous pituitary: at 20 days the ERß+ population was 10.7 ± 2.2%, while after 40 and 60 days of treatment an almost complete loss in the ERß expression was observed (40 d: 1 ± 0.6%; 60 d: 2 ± 0.6%). The ERα/ß ratio increased starting from tumors at 40 days, mainly due to the loss of ERß expression. The cell proliferation was analyzed in normal and hyperplastic pituitary and also in GH3ß- and GH3ß+ which contained different levels of ERß expression, and therefore different ERα/ß ratios. The over-expression of ERß inhibited the GH3 cell proliferation and expression of cyclin D1 and ERα. Also, the ERß activation by its agonist DPN changed the subcellular localization of p21, inducing an increase in the p21 nuclear expression, where it acts as a tumoral suppressor. These results show that ERß exerts an inhibitory role on pituitary cell proliferation, and that this effect may be partially due to the modulation of some key regulators of the cell cycle, such as cyclin D1 and p21. These data contribute significantly to the understanding of the ER effects in the proliferative control of pituitary gland, specifically related to the ERß function in the E2 actions on this endocrine gland.


Subject(s)
Cell Cycle/drug effects , Estradiol/adverse effects , Estrogen Receptor beta/metabolism , Pituitary Gland, Anterior/cytology , Pituitary Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Female , Gene Expression Regulation/drug effects , NAD/pharmacology , Pituitary Gland, Anterior/metabolism , Pituitary Neoplasms/chemically induced , Pituitary Neoplasms/pathology , Rats
8.
Int J Pharm ; 478(1): 258-267, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25448587

ABSTRACT

Large oral doses of ACZ lower the intraocular pressure (IOP), but usually lead to a multitude of systemic side effects, including gastrointestinal upset. The present study was undertaken to evaluate the effect of ACZ on the histological structure of rat duodenal mucosa and to assess a possible protective role of the complex formation of ACZ with HP-ß-CD, either separately or in combination with a third compound, on the gut epithelial layer by histological and ultrastructural examinations of sections of rat duodenum exposed to ACZ or its formulations. In addition, the transport process of ACZ and its binary or ternary complexes across the duodenal mucosa by means of the single-pass intestinal perfusion (SPIP) method in rats was evaluated. Evidence was found that ACZ alters intestinal permeability and induces damage to the rat small intestine. In contrast, ACZ-induced intestinal injury may be abrogated by ACZ complexation. In addition, the complexation of ACZ with HP-ß-CD, alone or in combination with a third compound, facilitated significant levels of ACZ uptake across the rat duodenal segment. Ternary complexes of ACZ with HP-ß-CD in combination with TEA (triethanolamine) or calcium ions were found to provide an excellent approach that enabled an increased apparent permeability of ACZ across the duodenal epithelium, with a concomitant ability to preserve the integrity of the gut epithelium from ACZ-induced injury. These results could be useful for the design and development of novel ACZ formulations that can reduce GI toxicity, while still maintaining their essential therapeutic efficacies.


Subject(s)
Acetazolamide , beta-Cyclodextrins , 2-Hydroxypropyl-beta-cyclodextrin , Acetazolamide/administration & dosage , Acetazolamide/chemistry , Acetazolamide/pharmacokinetics , Acetazolamide/toxicity , Animals , Calcium/administration & dosage , Calcium/chemistry , Calcium/pharmacokinetics , Calcium/toxicity , Duodenum/drug effects , Duodenum/pathology , Duodenum/ultrastructure , Ethanolamines/administration & dosage , Ethanolamines/chemistry , Ethanolamines/pharmacokinetics , Ethanolamines/toxicity , Intestinal Absorption , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestinal Mucosa/ultrastructure , Male , Microscopy, Electron, Transmission , Rats, Wistar , beta-Cyclodextrins/administration & dosage , beta-Cyclodextrins/chemistry , beta-Cyclodextrins/pharmacokinetics , beta-Cyclodextrins/toxicity
9.
Am J Physiol Endocrinol Metab ; 305(1): E41-9, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23651845

ABSTRACT

In the present work, we investigated the effect of 17ß-estradiol (E2) and basic fibroblast growth factor 2 (FGF2) on the lactotroph cell-proliferative response and the related membrane-initiated signaling pathway. Anterior pituitary mixed-cell cultures of random, cycling 3-mo-old female rats were treated with 10 nM E2, E2 membrane-impermeable conjugated BSA (E2-BSA), PPT (ERα agonist), and DPN (ERß agonist) alone or combined with FGF2 (10 ng/ml) for 30 min or 4 h. Although our results showed that the uptake of BrdU into the nucleus of lactotrophs was not modified by E2 or FGF2 alone, a significant increase in the lactotroph uptake of BrdU was observed after E2/FGF2 coincubation, with this effect being mimicked by PPT/FGF2. These proliferative effects were blocked by ICI 182,780 or PD-98059. The involvement of membrane ER in the proliferative response of prolactin cells induced by the steroid and FGF2 coincubation was confirmed using E2-BSA, and the association between ERα and FGF receptor was observed after E2/FGF2 treatment by immunoprecipitation. A significant increase in the ERK1/2 expression was noted after E2, E2-BSA, PPT, and FGF2 alone, which was more noticeable after E2-BSA/FGF2, E2/FGF2, or PPT/FGF2 treatments. This study provides evidence that E2 and FGF2 exert a cooperative effect on the lactotroph proliferation principally by signaling initiated at the plasma membrane triggering a genomic effect mediated by MEK/ERK1/2, a common signaling pathway, that finally regulates the lactotroph population, thus contributing to pituitary plasticity.


Subject(s)
Cell Membrane/metabolism , Estradiol/metabolism , Fibroblast Growth Factor 2/metabolism , Lactotrophs/metabolism , MAP Kinase Signaling System/physiology , Pituitary Gland, Anterior/metabolism , Animals , Cell Proliferation/drug effects , Drug Synergism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Female , Fibroblast Growth Factor 2/pharmacology , Lactotrophs/cytology , MAP Kinase Signaling System/drug effects , Pituitary Gland, Anterior/cytology , Primary Cell Culture , Rats , Rats, Wistar
14.
Am J Physiol Endocrinol Metab ; 302(10): E1189-97, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22354782

ABSTRACT

Considering that estradiol is a major modulator of prolactin (PRL) secretion, the aim of the present study was to analyze the role of membrane estradiol receptor-α (mERα) in the regulatory effect of this hormone on the PRL secretion induced by thyrotropin-releasing hormone (TRH) by focusing on the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) pathway activation. Anterior pituitary cell cultures from female rats were treated with 17ß-estradiol (E(2), 10 nM) and its membrane-impermeable conjugated estradiol (E(2)-BSA, 10 nM) alone or coincubated with TRH (10 nM) for 30 min, with PRL levels being determined by RIA. Although E(2), E(2)-BSA, TRH, and E(2)/TRH differentially increased the PRL secretion, the highest levels were achieved with E(2)-BSA/TRH. ICI-182,780 did not modify the TRH-induced PRL release but significantly inhibited the PRL secretion promoted by E(2) or E(2)-BSA alone or in coincubation with TRH. The PI3K inhibitors LY-294002 and wortmannin partially inhibited the PRL release induced by E(2)-BSA, TRH, and E(2)/TRH and totally inhibited the PRL levels stimulated by E(2)-BSA/TRH, suggesting that the mER mediated the cooperative effect of E(2) on TRH-induced PRL release through the PI3K pathway. Also, the involvement of this kinase was supported by the translocation of its regulatory subunit p85α from the cytoplasm to the plasma membrane in the lactotroph cells treated with E(2)-BSA and TRH alone or in coincubation. A significant increase of phosphorylated Akt was induced by E(2)-BSA/TRH. Finally, the changes of ERα expression in the plasmalemma of pituitary cells were examined by confocal microscopy and flow cytometry, which revealed that the mobilization of intracellular ERα to the plasma membrane of lactotroph cells was only induced by E(2). These finding showed that E(2) may act as a modulator of the secretory response of lactotrophs induced by TRH through mER, with the contribution by PI3K/Akt pathway activation providing a new insight into the mechanisms underlying the nongenomic action of E(2) in the pituitary.


Subject(s)
Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Pituitary Gland, Anterior , Prolactin/metabolism , Signal Transduction/physiology , Thyrotropin-Releasing Hormone/metabolism , Animals , Cell Membrane/metabolism , Cells, Cultured , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Female , Membrane Proteins/metabolism , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/drug effects , Pituitary Gland, Anterior/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Serum Albumin, Bovine/pharmacology , Signal Transduction/drug effects
15.
Mol Cell Endocrinol ; 355(1): 169-79, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22366173

ABSTRACT

In the present work we aimed at identifying ERα in the plasma membrane of normal anterior pituitary cells and investigated if 17ß-estradiol was able to induce their subcellular redistribution. Our results show that about 8% of anterior pituitary cells expressed ERα in the plasma membrane, with the geometrical mean fluorescence intensity being increased after steroid hormone treatment. 17ß-Estradiol and the selective ERα agonist PPT induced an increase of ERα expression in the plasma membrane and activated the PKCα/ERK 1/2 pathway in a time-course not compatible with genomic actions, thus supporting the notion of membrane-initiated effects. These findings suggest that 17ß-estradiol stimulates the translocation of endogenous ERα to the plasma membrane, consequently modulating this ER pool and leading to cellular biological effects in normal anterior pituitary gland.


Subject(s)
Cell Membrane/drug effects , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Pituitary Gland, Anterior/drug effects , Animals , Cell Membrane/metabolism , Cells, Cultured , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression/drug effects , Ligands , Phenols , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Transport/drug effects , Pyrazoles/pharmacology , Rats , Rats, Wistar , Signal Transduction/drug effects , Up-Regulation
16.
Ann Clin Microbiol Antimicrob ; 10: 15, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21527033

ABSTRACT

BACKGROUND: Community-Associated Methicillin Resistant Staphylococcus aureus (CA-MRSA) has traditionally been related to skin and soft tissue infections in healthy young patients. However, it has now emerged as responsible for severe infections worldwide, for which vancomycin is one of the mainstays of treatment. Infective endocarditis (IE) due to CA-MRSA with heterogeneous vancomycin-intermediate susceptibility-(h-VISA) has been recently reported, associated to an epidemic USA 300 CA-MRSA clone. CASE PRESENTATION: We describe the occurrence of h-VISA phenotype in a case of IE caused by a strain belonging to an epidemic CA-MRSA clone, distinct from USA300, for the first time in Argentina. The isolate h-VISA (SaB2) was recovered from a patient with persistent bacteraemia after a 7-day therapy with vancomycin, which evolved to fatal case of IE complicated with brain abscesses. The initial isolate-(SaB1) was fully vancomycin susceptible (VSSA). Although MRSA SaB2 was vancomycin susceptible (≤ 2 µg/ml) by MIC (agar and broth dilution, E-test and VITEK 2), a slight increase of MIC values between SaB1 and SaB2 isolates was detected by the four MIC methods, particularly for teicoplanin. Moreover, Sab2 was classified as h-VISA by three different screening methods [MHA5T-screening agar, Macromethod-E-test-(MET) and by GRD E-test] and confirmed by population analysis profile-(PAP). In addition, a significant increase in cell-wall thickness was revealed for SaB2 by electron microscopy. Molecular typing showed that both strains, SaB1 and SaB2, belonged to ST5 lineage, carried SCCmecIV, lacked Panton-Valentine leukocidin-(PVL) genes and had indistinguishable PFGE patterns (subtype I2), thereby confirming their isogenic nature. In addition, they were clonally related to the epidemic CA-MRSA clone (pulsotype I) detected in our country. CONCLUSIONS: This report demonstrates the ability of this epidemic CA-MRSA clone, disseminated in some regions of Argentina, to produce severe and rapidly fatal infections such as IE, in addition to its ability to acquire low-level vancomycin resistance; for these reasons, it constitutes a new challenge for the Healthcare System of this country.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Endocarditis, Bacterial/microbiology , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Staphylococcal Infections/microbiology , Vancomycin/therapeutic use , Aged , Argentina/epidemiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Endocarditis, Bacterial/drug therapy , Endocarditis, Bacterial/epidemiology , Epidemics , Female , Humans , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/metabolism , Staphylococcal Infections/drug therapy , Staphylococcal Infections/epidemiology , Vancomycin Resistance
17.
Exp Physiol ; 96(2): 226-39, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21075822

ABSTRACT

Lactotroph cells display morphological and functional heterogeneity, a feature which is closely related to the oestrogenic environment. In this study, we focused on sex-related differences linked to the proliferative and secretory responses of lactotrophs exposed to EGF in vitro. Furthermore, we addressed the involvement of the PKCε/ERK1/2 signalling pathway and the contribution of Pit-1 in the EGF actions in primary pituitary cultures from male and female rats. EGF promoted a differential proliferative activity on PRL cells, which was closely associated to the sex, as revealed by the uptake of bromodeoxyuridine (BrdU). In females, the mitogenic activity was up to nine times greater, whereas in males, the number of BrdU-labelled PRL cells was only doubled compared to control. However, in both models, EGF had a similar effectiveness in promoting PRL secretion. EGF also induced a significant increase in the PKCε, P -ERK 1/2, and Pit-1 protein levels, which were higher in females than in males. Pre-incubation with BIM blocked EGF-induced ERK 1/2 activation and Pit-1 expression. These results suggest a sexually dimorphic response of lactotroph cells to the proliferative effects of EGF, with the PKCε/ERK1/2 Pit-1 pathway being involved in this action.


Subject(s)
Cell Proliferation/drug effects , Epidermal Growth Factor/pharmacology , Lactotrophs/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Kinase C-epsilon/metabolism , Transcription Factor Pit-1/metabolism , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Female , Lactotrophs/enzymology , Lactotrophs/metabolism , Male , Phosphorylation , Prolactin/metabolism , Rats, Wistar , Sex Characteristics , Sex Factors , Signal Transduction/drug effects
18.
Acta Neuropathol ; 112(4): 491-501, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16823503

ABSTRACT

Fibroblast growth factor-2 (FGF-2) synthesized in the pituitary is involved in the formation and progression of pituitary tumors. The aim of this study was to analyze the pattern expression of two FGF-2 isoforms at different subcellular levels and to determine its correlation with prolactinoma development. Estrogen administration to male rats for 7, 20, and 60 days generated pituitary tumors, with lactotrophs being the prevalent cell type. Ultrastructural immunolabeling showed FGF-2 in the cytosolic and nuclear compartments of somatotrophs, lactotrophs and gonadotrophs, as well as in folliculo-stellate cells of normal rats. Estrogen stimulation increased FGF-2 immunoreactivity in various tumors and enhanced the expression of two FGF-2 isoforms, 18 and 22 kDa, as quantified by western blot. The 18 kDa isoform observed in cytosol extracts reached the highest levels after 60 days of hormonal stimulation and this was related to lactotroph proliferation. However, the 22 kDa FGF-2 isoform was only detected in the nuclear compartment and achieved the maximum expression at 7 days of estrogen treatment, without any correlation with lactotroph proliferation. These results suggest that the 18 kDa FGF-2 may play a role in the modulation of lactotroph proliferation in prolactinomas induced by estrogen. The overproduction of both FGF-2 isoforms appears to be implicated in autocrine-paracrine-intracrine mitogenic loops; this FGF-2 activity could lead to uncontrolled cell growth, angiogenesis, and tumor formation.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation, Neoplastic/physiology , Pituitary Neoplasms/metabolism , Prolactinoma/metabolism , Animals , Blotting, Western/methods , Disease Models, Animal , Estradiol/analogs & derivatives , Fibroblast Growth Factor 2/genetics , Gene Expression Regulation, Neoplastic/drug effects , Immunohistochemistry/methods , Male , Microscopy, Electron, Transmission/methods , Molecular Weight , Pituitary Neoplasms/chemically induced , Pituitary Neoplasms/ultrastructure , Prolactin/metabolism , Prolactinoma/chemically induced , Prolactinoma/ultrastructure , Protein Isoforms/genetics , Protein Isoforms/metabolism , Radioimmunoassay/methods , Rats , Rats, Wistar , Reticulin/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...