Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Clin Invest ; 118(6): 2157-68, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18451999

RESUMO

Paroxysmal dyskinesias are episodic movement disorders that can be inherited or are sporadic in nature. The pathophysiology underlying these disorders remains largely unknown but may involve disrupted ion homeostasis due to defects in cell-surface channels or nutrient transporters. In this study, we describe a family with paroxysmal exertion-induced dyskinesia (PED) over 3 generations. Their PED was accompanied by epilepsy, mild developmental delay, reduced CSF glucose levels, hemolytic anemia with echinocytosis, and altered erythrocyte ion concentrations. Using a candidate gene approach, we identified a causative deletion of 4 highly conserved amino acids (Q282_S285del) in the pore region of the glucose transporter 1 (GLUT1). Functional studies in Xenopus oocytes and human erythrocytes revealed that this mutation decreased glucose transport and caused a cation leak that alters intracellular concentrations of sodium, potassium, and calcium. We screened 4 additional families, in which PED is combined with epilepsy, developmental delay, or migraine, but not with hemolysis or echinocytosis, and identified 2 additional GLUT1 mutations (A275T, G314S) that decreased glucose transport but did not affect cation permeability. Combining these data with brain imaging studies, we propose that the dyskinesias result from an exertion-induced energy deficit that may cause episodic dysfunction of the basal ganglia, and that the hemolysis with echinocytosis may result from alterations in intracellular electrolytes caused by a cation leak through mutant GLUT1.


Assuntos
Anemia Hemolítica/etiologia , Anemia Hemolítica/genética , Cátions , Coreia/genética , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/fisiologia , Glucose/metabolismo , Adulto , Sequência de Aminoácidos , Animais , Coreia/patologia , Eritrócitos/metabolismo , Feminino , Humanos , Masculino , Modelos Biológicos , Dados de Sequência Molecular , Esforço Físico , Xenopus
2.
Cell Physiol Biochem ; 23(1-3): 191-8, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19255513

RESUMO

Paclitaxel triggers suicidal erythrocyte death or eryptosis, characterized by exposure of phosphatidylserine at the erythrocyte surface and cell shrinkage. Eryptosis of infected erythrocytes may delay development of parasitemia and thus favourably influence the course of malaria. The present study explored whether paclitaxel influences in vitro parasite growth and eryptosis of Plasmodium falciparum infected human erythrocytes and in vivo parasitemia and survival of Plasmodium berghei infected mice. Phosphatidylserine exposing erythrocytes were identified utilizing annexin V binding and erythrocyte volume was estimated from forward scatter in FACS analysis. In vitro infection of human erythrocytes with P. falciparum increased annexin binding and decreased forward scatter, effects augmented in the presence of paclitaxel (> or = 0.01 microM). Paclitaxel (> or = 0.01 microM) significantly decreased intraerythrocytic DNA/RNA content and in vitro parasitemia. In Plasmodium berghei infected mice parasitemia was significantly decreased (from 55.8% to 28.6% of circulating erythrocytes 20 days after infection) and mouse survival significantly enhanced (from 0% to 69.23% 25 days after infection) by administration of 8.5 mg/kg.b.w. of paclitaxel intraperitoneally from the eighth day of infection. In conclusion, paclitaxel decreases parasitemia and enhances survival of P. berghei infected mice, an effect, which may be due to stimulation of eryptosis and/or a direct toxic effect on the parasite.


Assuntos
Antimaláricos/farmacologia , Eritrócitos/efeitos dos fármacos , Eritrócitos/parasitologia , Malária/tratamento farmacológico , Paclitaxel/farmacologia , Plasmodium berghei/efeitos dos fármacos , Plasmodium berghei/fisiologia , Animais , Anexina A5/metabolismo , Antimaláricos/uso terapêutico , Morte Celular/efeitos dos fármacos , Células Cultivadas , Eritrócitos/metabolismo , Feminino , Citometria de Fluxo , Humanos , Malária/mortalidade , Masculino , Camundongos , Paclitaxel/uso terapêutico , Parasitemia/tratamento farmacológico , Parasitemia/mortalidade
3.
Cell Physiol Biochem ; 24(5-6): 415-28, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19910682

RESUMO

The course of malaria does not only depend on the virulence of the parasite Plasmodium but also on properties of host erythrocytes. Here, we show that infection of erythrocytes from human sickle cell trait (HbA/S) carriers with ring stages of P. falciparum led to significantly enhanced PGE(2) formation, Ca(2+) permeability, annexin-A7 degradation, phosphatidylserine (PS) exposure at the cell surface, and clearance by macrophages. P. berghei-infected erythrocytes from annexin-A7-deficient (annexin-A7(-/-)) mice were more rapidly cleared than infected wildtype cells. Accordingly, P. berghei-infected annexin-A7(-/-) mice developed less parasitemia than wildtype mice. The cyclooxygenase inhibitor aspirin decreased erythrocyte PS exposure in infected annexin-A7(-/-) mice and abolished the differences of parasitemia and survival between the genotypes. Conversely, the PGE(2)-agonist sulprostone decreased parasitemia and increased survival of wild type mice. In conclusion, PS exposure on erythrocytes results in accelerated clearance of Plasmodium ring stage-infected HbA/S or annexin-A7(-/-) erythrocytes and thus confers partial protection against malaria in vivo.


Assuntos
Anexina A7/metabolismo , Eritrócitos/metabolismo , Plasmodium falciparum/fisiologia , Traço Falciforme/parasitologia , Animais , Anexina A7/deficiência , Anexina A7/genética , Aspirina/uso terapêutico , Cálcio/metabolismo , Dinoprostona/análogos & derivados , Dinoprostona/metabolismo , Dinoprostona/uso terapêutico , Eritrócitos/parasitologia , Genótipo , Hemoglobina A/metabolismo , Hemoglobina Falciforme/metabolismo , Humanos , Camundongos , Camundongos Knockout , Parasitemia/tratamento farmacológico , Fagocitose , Fosfatidilserinas/metabolismo , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium berghei/fisiologia , Plasmodium falciparum/crescimento & desenvolvimento , Traço Falciforme/metabolismo
4.
Cell Physiol Biochem ; 22(5-6): 405-12, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19088422

RESUMO

Plasmodia express a sphingomyelinase, which is apparently required for their development. On the other hand, the sphingomyelinase product ceramide has previously been shown to delay parasite development. Moreover, ceramide triggers suicidal erythrocyte death or eryptosis, characterized by exposure of phosphatidylserine at the erythrocyte surface and cell shrinkage. Accelerated eryptosis of infected erythrocytes is considered to clear infected erythrocytes from circulating blood and, thus, to favourably influence the clinical course of malaria. The present experiments explored whether the sphingomyelinase inhibitor amitriptyline or genetic knockout of host acid sphingomyelinase influence in vitro parasite growth, eryptosis of Plasmodium falciparum-infected human erythrocytes, in vivo parasitemia and survival of P. berghei-infected mice. Phosphatidylserine exposure was determined by annexin V-binding and cell volume by forward scatter in FACS analysis. In vitro infection of human erythrocytes increased annexin- binding, an effect blunted in the presence of amitriptyline (>or=50 microM). Amitriptyline did not significantly alter intraerythrocytic parasite development but significantly (>or= 1 microM) delayed the increase in parasitemia in vitro. Most importantly, amitriptyline treatment (1 mM in drinking water) resulted in a significant delay of parasitemia and death of infected mice. However, upon infection, ceramide formation was stimulated in both, acid sphingomyelinase knockout mice (Smpd1(-/-)) and their wild type littermates (Smpd1(+/+)). Parasitemia following P. berghei infection was significantly lower in Smpd1(-/-) than in Smpd1(+/+) mice but did not significantly extend the life span of infected animals. In conclusion, mammalian and parasite sphingomyelinase contribute to ceramide formation during malaria, whereby the parasite sphingomyelinase ultimately determines the course of the infection. Amitriptyline presumably blocks both sphingomyelinases and, thus, its use might be a novel strategy to treat malaria.


Assuntos
Amitriptilina/farmacologia , Apoptose/efeitos dos fármacos , Eritrócitos/citologia , Eritrócitos/parasitologia , Parasitemia/parasitologia , Plasmodium berghei/fisiologia , Animais , Ceramidas/biossíntese , Eritrócitos/efeitos dos fármacos , Feminino , Humanos , Malária/parasitologia , Masculino , Camundongos , Fosfatidilserinas/metabolismo , Plasmodium berghei/efeitos dos fármacos , Plasmodium berghei/crescimento & desenvolvimento , Análise de Sobrevida
5.
Cell Physiol Biochem ; 22(1-4): 261-8, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18769053

RESUMO

Chlorpromazine has previously been shown to trigger suicidal erythrocyte death or eryptosis, which is characterized by exposure of phosphatidylserine at the erythrocyte surface and cell shrinkage. Premature suicidal death of infected erythrocytes is in turn considered to delay development of parasitemia and thus favourably influence the clinical course of malaria. The present experiments have been performed to explore whether chlorpromazine influences in vitro parasite growth and eryptosis of Plasmodium falciparum infected human erythrocytes and in vivo parasitemia and survival of P. berghei infected mice. Phosphatidylserine was estimated from annexin V binding and cell volume from forward scatter in FACS analysis. In vitro infection of human erythrocytes increased annexin binding and decreased forward scatter, effects augmented in the presence of chlorpromazine (>or=10 microM). Chlorpromazine did not significantly alter intraerythrocytic DNA/RNA content but significantly (>or=1 microM) decreased in vitro parasitemia. In chlorpromazine treated mice erythrocytes were more rapidly cleared from circulating blood than in nontreated mice. Parasitemia in P. berghei infected mice was significantly decreased (from 50 % to 28 % of circulating erythrocytes 22 days after infection) and mouse survival significantly enhanced (from 0 % to 80 % 30 days after infection) by addition of 1 mM chlorpromazine to the drinking water from the first day of infection. In conclusion, chlorpromazine favourably influences the course of malaria, an effect at least partially due to stimulation of suicidal erythrocyte death.


Assuntos
Apoptose/efeitos dos fármacos , Clorpromazina/farmacologia , Eritrócitos/citologia , Eritrócitos/efeitos dos fármacos , Parasitemia/patologia , Parasitemia/parasitologia , Plasmodium berghei/fisiologia , Animais , Eritrócitos/parasitologia , Feminino , Humanos , Malária/parasitologia , Masculino , Camundongos , Fosfatidilserinas/metabolismo , Plasmodium berghei/efeitos dos fármacos , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/crescimento & desenvolvimento , Análise de Sobrevida
6.
Cell Physiol Biochem ; 21(5-6): 481-8, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18453756

RESUMO

Accelerated suicidal death or eryptosis of infected erythrocytes may delay development of parasitemia in malaria. Eryptosis is inhibited by nitric oxide (NO). The present study has been performed to explore, whether inhibition of NO synthase by L-NAME modifies the course of malaria. We show here that L-NAME (>or=10 microM) increased phosphatidylserine exposure of Plasmodium falciparum infected human erythrocytes, an effect significantly more marked than in noninfected human erythrocytes. We further show that parasitemia in Plasmodium berghei infected mice was significantly decreased (from 50% to 18% of circulating erythrocytes 20 days after infection) by addition of 1 mg/ml L-NAME to the drinking water. According to CFSE labelling L-NAME treatment accelerated the clearance of both, noninfected and infected, erythrocytes from circulating blood, but did not significantly extend the life span of infected animals. In conclusion, treatment with L-NAME shortens the life span of circulating erythrocytes and thus delays development of parasitemia during malaria.


Assuntos
NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Parasitemia/tratamento farmacológico , Plasmodium berghei/efeitos dos fármacos , Animais , Eritrócitos/efeitos dos fármacos , Feminino , Masculino , Camundongos , NG-Nitroarginina Metil Éster/uso terapêutico , Óxido Nítrico Sintase/metabolismo , Parasitemia/enzimologia , Taxa de Sobrevida
7.
Cell Physiol Biochem ; 22(5-6): 395-404, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19088421

RESUMO

The intraerythrocytic development of P. falciparum induces New Permeability Pathways (NPP) in the membrane of the parasitized erythrocyte which provide the parasite with nutrients, adjust the erythrocyte electrolyte composition to the needs of the parasite, and dispose of metabolic waste products and osmolytes. Patch-clamp recordings identified inwardly and outwardly rectifying (OR) anion conductances in the host erythrocyte membrane as electrophysiological correlate of the NPP. The OR conductance is regulated by serum. Here we show that serum albumin (SA) stimulated OR-generated Cl(-) and lactate outward currents with an EC(50) of approximately 100 nM while other proteins such as ovalbumin or casein did not. The stimulatory efficacy did not differ between fatty acid free bovine SA and recombinant human SA and disruption of the SA tertiary structure abolished the effect suggesting that intact SA protein and not other bound factors interact with the erythrocyte membrane. Taken together, the data indicate a high affinity and specificity interaction of native SA with the parasitized erythrocytes which might underlie the observed dependence of P. falciparum growth in vitro on SA.


Assuntos
Eritrócitos/metabolismo , Eritrócitos/parasitologia , Ativação do Canal Iônico , Malária/metabolismo , Plasmodium falciparum/metabolismo , Albumina Sérica/metabolismo , Animais , Caseínas/farmacologia , Canais de Cloreto/metabolismo , Eritrócitos/efeitos dos fármacos , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ácido Láctico/metabolismo , Ovalbumina/farmacologia , Plasmodium falciparum/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Soroalbumina Bovina/farmacologia
8.
Cell Physiol Biochem ; 22(1-4): 169-76, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18769043

RESUMO

Stimulation of the mast cell IgE-receptor (FcepsilonRI) by antigen leads to stimulation of Ca(2+) entry with subsequent mast cell degranulation and release of inflammatory mediators. Ca(2+) further activates Ca(2+)-activated K(+) channels, which in turn provide the electrical driving force for Ca(2+) entry. Since phosphatidylinositol (PI)-3-kinase has previously been shown to be required for mast cell activation and degranulation, we explored, whether mast cell Ca(2+) and Ca(2+)-activated K(+) channels may be sensitive to PI3-kinase activity. Whole-cell patch clamp experiments and Fura-2 fluorescence measurements for determination of cytosolic Ca(2+) concentration were performed in mouse bone marrow-derived mast cells either treated or untreated with the PI3-kinase inhibitors LY-294002 (10 muM) and wortmannin (100 nM). Antigen-stimulated Ca(2+) entry but not Ca(2+) release from the intracellular stores was dramatically reduced upon PI3-kinase inhibition. Ca(2+) entry was further inhibited by TRPV blocker ruthenium red (10 muM). Ca(2+) entry following readdition after Ca(+)-store depletion with thapsigargin was again decreased by LY-294002, pointing to inhibition of store-operated channels (SOCs). Moreover, inhibition of PI3-kinase abrogated IgE-stimulated, but not ionomycin-induced stimulation of Ca(2+)-activated K(+) channels. These observations disclose PI3-kinase-dependent regulation of Ca(2+) entry and Ca(2+)-activated K(+)-channels, which in turn participate in triggering mast cell degranulation.


Assuntos
Ativação do Canal Iônico , Mastócitos/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Animais , Antígenos/farmacologia , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Canais de Cálcio/metabolismo , Degranulação Celular/efeitos dos fármacos , Cromonas/farmacologia , Feminino , Hexosaminidases/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Masculino , Mastócitos/efeitos dos fármacos , Mastócitos/fisiologia , Camundongos , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Canais de Potássio Cálcio-Ativados/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Rutênio Vermelho/farmacologia
9.
Biochem Biophys Res Commun ; 376(3): 514-8, 2008 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-18804454

RESUMO

Infection of human erythrocytes with the malaria parasite Plasmodium falciparum induces activation of organic osmolyte and anion channels in the host cell membrane. These channels supply the intraerythrocytic parasite with nutrients, dispose of metabolic waste products, adjust the host electrolyte concentrations to the parasite's needs, and lower the colloid osmotic pressure, thus preventing premature hemolysis of the osmotically challenged host cell. Four different types of anion channels (CFTR, ClC-2 or PSAC, an 18pS inward rectifier, and an 80pS outward rectifier) have been identified in human erythrocytes. Here, we show that the 80pS channels underlie a serum albumin-dependent anion current. Both, the parasite in vitro development and the organic osmolyte permeability of the parasitized erythrocyte, reportedly depend on serum albumin, highlighting the pivotal functional significance of the 80pS channel for the intraerythrocytic parasite development.


Assuntos
Eritrócitos/metabolismo , Eritrócitos/parasitologia , Canais Iônicos/metabolismo , Malária Falciparum/metabolismo , Plasmodium falciparum , Albumina Sérica/metabolismo , Animais , Células Cultivadas , Membrana Eritrocítica/efeitos dos fármacos , Membrana Eritrocítica/metabolismo , Membrana Eritrocítica/parasitologia , Eritrócitos/efeitos dos fármacos , Humanos , Nitrobenzoatos/farmacologia
10.
Biochem Biophys Res Commun ; 376(3): 494-8, 2008 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-18789889

RESUMO

Cyclosporine triggers suicidal erythrocyte death or eryptosis, which is characterized by cell shrinkage and exposure of phosphatidylserine at the erythrocyte surface. The present study explored whether cyclosporine influences eryptosis of Plasmodium infected erythrocytes, development of parasitemia and thus the course of the disease. Annexin V binding was utilized to depict phosphatidylserine exposure and forward scatter in FACS analysis to estimate erythrocyte volume. In vitro infection of human erythrocytes with Plasmodium falciparum increased annexin binding and decreased forward scatter, effects potentiated by cyclosporine (> or = 0.01 microM). Cyclosporine (> or = 0.001 microM) significantly decreased intraerythrocytic DNA/RNA content and in vitro parasitemia (> or = 0.01 microM). Administration of cyclosporine (5 mg/kg b.w.) subcutaneously significantly decreased the parasitemia (from 47% to 27% of circulating erythrocytes 20 days after infection) and increased the survival of P. berghei infected mice (from 0% to 94% 30 days after infection). In conclusion, cyclosporine augments eryptosis, decreases parasitemia and enhances host survival during malaria.


Assuntos
Apoptose/efeitos dos fármacos , Ciclosporina/farmacologia , Eritrócitos/efeitos dos fármacos , Malária/parasitologia , Parasitemia/parasitologia , Plasmodium berghei/efeitos dos fármacos , Animais , Membrana Eritrocítica/efeitos dos fármacos , Membrana Eritrocítica/metabolismo , Membrana Eritrocítica/parasitologia , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Eritrócitos/patologia , Camundongos , Camundongos Endogâmicos , Fosfatidilserinas/metabolismo , Plasmodium berghei/fisiologia
11.
Anatol J Cardiol ; 19(5): 326-323, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29724975

RESUMO

OBJECTIVE: Energy drinks (EDs) target young and active individuals and they are being marketed as enhancers of energy, concentration, and physical and cognitive performance. Their long-term consumption raises serious health concerns related to cardiovascular events. Here we investigate the effects of long-term Red Bull® consumption and its combination with alcohol on certain biochemical parameters and the ultrastructure of the myocardium. METHODS: Male Wistar rats were categorized into four groups and given different treatments via oral administration. The Control (C) group received tap water, the Red Bull (RB) group received 1.5 ml/100 g body weight of Red Bull, the ethanol group (E) received 0.486 mg/100 g body weight of ethanol, and the Red Bull and ethanol (RBE) received a combination of the two beverages for 30 days. In the last 6 days of the experiment, the animals were tested for their physical performance by conducting a weight-loaded forced swim test. Immediately after swimming exhaustion, the animals were sacrificed under anesthesia and samples of the heart muscle were harvested for ultrastructural and biochemical analyses. RESULTS: Our results showed a significant increase in the heart glucose and glycogen concentrations in the RB and RBE groups. Total cholesterol concentration significantly decreased in the RBE and RB groups. Total protein concentration and ALT and AST activities increased in all groups. The biochemical changes were accompanied by ultrastructural alterations. CONCLUSION: Based on these results, we recommend that athletes and active persons should avoid the long-term consumption of the Red Bull ED and, particularly, its combination with alcohol.


Assuntos
Estimulantes do Sistema Nervoso Central/farmacologia , Bebidas Energéticas/efeitos adversos , Miocárdio/patologia , Resistência Física/efeitos dos fármacos , Animais , Teste de Esforço , Masculino , Ratos , Ratos Wistar
12.
FEBS Lett ; 581(28): 5407-12, 2007 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-17976378

RESUMO

To test for redox regulation of anion channels in erythroid cells, we exposed K562 cells to oxidants and measured changes in transmembrane Cl(-) currents using patch-clamp, and in intracellular Cl(-) content using the Cl(-) selective dye MQAE. Oxidation with tert-butylhydroperoxide or H(2)O(2) produced a plasma membrane anion permeability with a permselectivity of NO(3)(-)>lactate(-)>gluconate(-). The permeability increase was paralleled by insertion of ClC-3 protein into the plasma membrane as evident from immunofluorescence microscopy and surface biotinylation. Down-regulation of ClC-3 protein by RNA interference as assessed by immunoblotting decreased the oxidation-stimulated permeability. In conclusion, oxidation induces surface expression of ClC-3 and activation of a ClC-3-dependent anion permeability in K562 cells.


Assuntos
Canais de Cloreto/metabolismo , Leucemia/metabolismo , Ânions/química , Linhagem Celular Tumoral , Canais de Cloreto/química , Canais de Cloreto/genética , Eletrofisiologia , Humanos , Leucemia/genética , Oxirredução , Técnicas de Patch-Clamp , RNA Interferente Pequeno/genética
13.
Cell Physiol Biochem ; 21(1-3): 183-92, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18209485

RESUMO

Human erythrocytes express cation channels which contribute to the background leak of Ca(2+), Na(+) and K(+). Excessive activation of these channels upon energy depletion, osmotic shock, Cl(-) depletion, or oxidative stress triggers suicidal death of erythrocytes (eryptosis), characterized by cell-shrinkage and exposure of phosphatidylserine at the cell surface. Eryptotic cells are supposed to be cleared from circulating blood. The present study aimed to identify the cation channels. RT-PCR revealed mRNA encoding the non-selective cation channel TRPC6 in erythroid progenitor cells. Western blotting indicated expression of TRPC6 protein in erythrocytes from man and wildtype mice but not from TRPC6(-/-) mice. According to flow-cytometry, Ca(2+) entry into human ghosts prepared by hemolysis in EGTA-buffered solution containing the Ca(2+) indicator Fluo3/AM was inhibited by the reducing agent dithiothreitol and the erythrocyte cation channel blockers ethylisopropylamiloride and amiloride. Loading of the ghosts with antibodies against TRPC6 or TRPC3/6/7 but neither with antibodies against TRPM2 or TRPC3 nor antibodies pre-adsorbed with the immunizing peptides inhibited ghost Ca(2+) entry. Moreover, free Ca(2+) concentration, cell-shrinkage, and phospholipid scrambling were significantly lower in Cl(-)-depleted TRPC6(-/-) erythrocytes than in wildtype mouse erythrocytes. In conclusion, human and mouse erythrocytes express TRPC6 cation channels which participate in cation leak and Ca(2+)-induced suicidal death.


Assuntos
Cálcio/metabolismo , Eritrócitos/metabolismo , Canais de Cátion TRPC/metabolismo , Animais , Citosol , Eritrócitos/citologia , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/metabolismo , Humanos , Ativação do Canal Iônico , Camundongos , Canal de Cátion TRPC6
14.
Cell Physiol Biochem ; 21(1-3): 193-202, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18209486

RESUMO

Vitamin A and retinoic acid have previously been shown to confer some protection against a severe course of malaria by fostering the phagocytosis of parasitized erythrocytes. Phagocytosis of erythrocytes is stimulated by phosphatidylserine exposure at the cell surface. The present study has thus been performed to explore the effect of retinoic acid and the specific retinoic acid receptor (RAR) agonist 4-(E-2-[5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl]-1-propenyl) benzoic acid (TTNPB) on erythrocyte annexin V binding, which reflects phosphatidylserine exposure at the cell surface. A 24 hours exposure to either, retinoic acid (3 microM) or TTNPB (3 microM), indeed significantly increased annexin binding, an effect paralleled by decrease of forward scatter reflecting cell shrinkage. According to Fluo3 fluorescence, exposure to either, retinoic acid (10 microM, 24 hours) or TTNPB (10 microM, 6 hours), significantly increased cytosolic Ca(2+)-activity, a known trigger of phosphatidylserine exposure. Infection of erythrocytes with Plasmodium falciparum increased phosphatidylserine exposure, an effect increased in the presence of TTNPB. In conclusion, retinoid acid and TTNPB trigger phosphatididylserine exposure and cell shrinkage of erythrocytes, typical features of suicidal erythrocyte death or eryptosis. The eryptosis could participate in the accelerated clearance of parasitized erythrocytes from circulating blood following treatment with retinoids.


Assuntos
Eritrócitos/citologia , Eritrócitos/efeitos dos fármacos , Tretinoína/farmacologia , Compostos de Anilina , Animais , Anexinas/metabolismo , Benzoatos/farmacologia , Calpaína/metabolismo , Caspases/metabolismo , Morte Celular/efeitos dos fármacos , Eritrócitos/enzimologia , Eritrócitos/microbiologia , Fluorescência , Glutationa/metabolismo , Humanos , Plasmodium falciparum , Ligação Proteica/efeitos dos fármacos , Retinoides/farmacologia , Xantenos
15.
Biochem Biophys Res Commun ; 357(3): 608-14, 2007 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-17445762

RESUMO

Iron deficiency accelerates suicidal erythrocyte death, which is evident from phosphatidylserine exposure. The present study explored whether iron deficiency compromises intraerythrocytic growth of Plasmodium and enhances death of infected erythrocytes thus influencing the course of malaria. As a result, phosphatidylserine exposure is increased in Plasmodium falciparum infected human erythrocytes, an effect significantly more marked in iron deficiency. Moreover, iron deficiency impairs in vitro intraerythrocytic growth and infection of erythrocytes. In mice, iron-deficient erythrocytes are more rapidly cleared from circulating blood, an effect increased by infection with Plasmodium berghei. Parasitemia in P. berghei infected mice was significantly decreased (from 54% to 33% of circulating erythrocytes 20 days after infection) and mouse survival significantly enhanced (from 0% to 20% 30 days after infection) in iron-deficient mice. In conclusion, iron deficiency favourably influences the course of malaria, an effect partially due to accelerated suicidal death and subsequent clearance of infected erythrocytes.


Assuntos
Eritrócitos/metabolismo , Deficiências de Ferro , Malária/metabolismo , Plasmodium berghei/crescimento & desenvolvimento , Análise de Variância , Animais , Eritrócitos/efeitos dos fármacos , Eritrócitos/parasitologia , Feminino , Citometria de Fluxo/métodos , Humanos , Malária/sangue , Malária/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilserinas/farmacologia , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/crescimento & desenvolvimento
16.
Biochem Biophys Res Commun ; 363(3): 484-9, 2007 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-17888870

RESUMO

Malaria, a disease accounting for more than one million deaths per year, is caused by intraerythrocytic growth of Plasmodia. Parasitemia may be blunted by suicidal erythrocyte death or eryptosis, which is characterized by cell shrinkage and phosphatidylserine exposure. Triggers of eryptosis include lead nitrate (Pb(NO3)2). As shown here, Pb(NO3)2 (> or = 10 microM) increased phosphatidylserine exposure of Plasmodium falciparum-infected human erythrocytes, an effect significantly more marked than in noninfected cells. Pb(NO3)2 treatment accelerated the clearance of erythrocytes from circulating blood. Parasitemia in Plasmodium berghei-infected mice was significantly decreased and mouse survival significantly enhanced by 100 microM Pb(NO3)2 (20 ppm) in drinking water. The treatment significantly increased reticulocyte number but did not significantly decrease erythrocyte number in noninfected mice and in infected animals mainly triggered the disappearance of P. berghei harbouring erythrocytes. In conclusion, Pb(NO3)2 accelerates eryptosis and subsequent clearance of infected erythrocytes and thus favourably influences the course of malaria.


Assuntos
Chumbo/farmacologia , Malária/prevenção & controle , Nitratos/farmacologia , Parasitemia/prevenção & controle , Plasmodium berghei/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Contagem de Eritrócitos , Eritrócitos/citologia , Eritrócitos/efeitos dos fármacos , Eritrócitos/parasitologia , Citometria de Fluxo , Humanos , Chumbo/uso terapêutico , Malária/sangue , Malária/parasitologia , Camundongos , Nitratos/uso terapêutico , Parasitemia/parasitologia , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/crescimento & desenvolvimento , Reticulócitos/citologia , Reticulócitos/efeitos dos fármacos , Análise de Sobrevida , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA