Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Biochimie ; 216: 3-13, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37820991

RESUMO

Alpha hemolysin (HlyA) is a hemolytic and cytotoxic protein secreted by uropathogenic strains of E. coli. The role of glycophorins (GPs) as putative receptors for HlyA binding to red blood cells (RBCs) has been debated. Experiments using anti-GPA/GPB antibodies and a GPA-specific epitope nanobody to block HlyA-GP binding on hRBCs, showed no effect on hemolytic activity. Similarly, the hemolysis induced by HlyA remained unaffected when hRBCs from a GPAnull/GPBnull variant were used. Surface Plasmon Resonance experiments revealed similar values of the dissociation constant between GPA and either HlyA, ProHlyA (inactive protoxin), HlyAΔ914-936 (mutant of HlyA lacking the binding domain to GPA) or human serum albumin, indicating that the binding between the proteins and GPA is not specific. Although far Western blot followed by mass spectroscopy analyses suggested that HlyA interacts with Band 3 and spectrins, hemolytic experiments on spectrin-depleted hRBCs and spherocytes, indicated these proteins do not mediate the hemolytic process. Our results unequivocally demonstrate that neither glycophorins, nor Band 3 and spectrins mediate the cytotoxic activity of HlyA on hRBCs, thereby challenging the HlyA-receptor hypothesis. This finding holds significant relevance for the design of anti-toxin therapeutic strategies, particularly in light of the growing antibiotic resistance exhibited by bacteria.


Assuntos
Proteínas de Escherichia coli , Toxinas Biológicas , Humanos , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas Hemolisinas/farmacologia , Proteínas Hemolisinas/química , Proteínas Hemolisinas/metabolismo , Proteínas de Membrana/metabolismo , Glicoforinas/metabolismo , Glicoforinas/farmacologia , Hemólise , Eritrócitos/metabolismo , Toxinas Biológicas/metabolismo
2.
Int J Mol Sci ; 23(16)2022 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-36012667

RESUMO

Pannexin 1 (PANX1) was proposed to drive ATP release from red blood cells (RBCs) in response to stress conditions. Stomatin, a membrane protein regulating mechanosensitive channels, has been proposed to modulate PANX1 activity in non-erythroid cells. To determine whether stomatin modulates PANX1 activity in an erythroid context, we have (i) assessed the in situ stomatin-PANX1 interaction in RBCs, (ii) measured PANX1-stimulated activity in RBCs expressing stomatin or from OverHydrated Hereditary Stomatocytosis (OHSt) patients lacking stomatin, and in erythroid K562 cells invalidated for stomatin. Proximity Ligation Assay coupled with flow imaging shows 27.09% and 6.13% positive events in control and OHSt RBCs, respectively. The uptake of dyes 5(6)-Carboxyfluorescein (CF) and TO-PRO-3 was used to evaluate PANX1 activity. RBC permeability for CF is 34% and 11.8% in control and OHSt RBCs, respectively. PANX1 permeability for TO-PRO-3 is 35.72% and 18.42% in K562 stom+ and stom- clones, respectively. These results suggest an interaction between PANX1 and stomatin in human RBCs and show a significant defect in PANX1 activity in the absence of stomatin. Based on these results, we propose that stomatin plays a major role in opening the PANX1 pore by being involved in a caspase-independent lifting of autoinhibition.


Assuntos
Desequilíbrio Ácido-Base , Conexinas , Eritrócitos , Proteínas de Membrana , Proteínas do Tecido Nervoso , Desequilíbrio Ácido-Base/metabolismo , Trifosfato de Adenosina/metabolismo , Anemia Hemolítica Congênita , Conexinas/metabolismo , Eritrócitos/metabolismo , Eritrócitos Anormais/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Erros Inatos do Metabolismo , Proteínas do Tecido Nervoso/metabolismo
3.
Int J Mol Sci ; 23(2)2022 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-35055067

RESUMO

α-hemolysin (HlyA) of E. coli binds irreversibly to human erythrocytes and induces cell swelling, ultimately leading to hemolysis. We characterized the mechanism involved in water transport induced by HlyA and analyzed how swelling and hemolysis might be coupled. Osmotic water permeability (Pf) was assessed by stopped-flow light scattering. Preincubation with HlyA strongly reduced Pf in control- and aquaporin 1-null red blood cells, although the relative Pf decrease was similar in both cell types. The dynamics of cell volume and hemolysis on RBCs was assessed by electrical impedance, light dispersion and hemoglobin release. Results show that HlyA induced erythrocyte swelling, which is enhanced by purinergic signaling, and is coupled to osmotic hemolysis. We propose a mathematical model of HlyA activity where the kinetics of cell volume and hemolysis in human erythrocytes depend on the flux of osmolytes across the membrane, and on the maximum volume that these cells can tolerate. Our results provide new insights for understanding signaling and cytotoxicity mediated by HlyA in erythrocytes.


Assuntos
Tamanho Celular , Eritrócitos/citologia , Eritrócitos/fisiologia , Proteínas de Escherichia coli/farmacologia , Proteínas Hemolisinas/farmacologia , Modelos Biológicos , Trifosfato de Adenosina/metabolismo , Biomarcadores , Morte Celular/efeitos dos fármacos , Morte Celular/imunologia , Relação Dose-Resposta a Droga , Proteínas de Escherichia coli/imunologia , Proteínas Hemolisinas/imunologia , Hemólise , Interações Hospedeiro-Patógeno , Humanos , Cinética , Permeabilidade
4.
J Biol Chem ; 298(1): 101503, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34929164

RESUMO

Hydrogen peroxide (H2O2) not only is an oxidant but also is an important signaling molecule in vascular biology, mediating several physiological functions. Red blood cells (RBCs) have been proposed to be the primary sink of H2O2 in the vasculature because they are the main cellular component of blood with a robust antioxidant defense and a high membrane permeability. However, the exact permeability of human RBC to H2O2 is neither known nor is it known if the mechanism of permeation involves the lipid fraction or protein channels. To gain insight into the permeability process, we measured the partition constant of H2O2 between water and octanol or hexadecane using a novel double-partition method. Our results indicated that there is a large thermodynamic barrier to H2O2 permeation. The permeability coefficient of H2O2 through phospholipid membranes containing cholesterol with saturated or unsaturated acyl chains was determined to be 4 × 10-4 and 5 × 10-3 cm s-1, respectively, at 37 °C. The permeability coefficient of human RBC membranes to H2O2 at 37 °C, on the other hand, was 1.6 × 10-3 cm s-1. Different aquaporin-1 and aquaporin-3 inhibitors proved to have no effect on the permeation of H2O2. Moreover, human RBCs devoid of either aquaporin-1 or aquaporin-3 were equally permeable to H2O2 as normal human RBCs. Therefore, these results indicate that H2O2 does not diffuse into RBCs through aquaporins but rather through the lipid fraction or a still unidentified membrane protein.


Assuntos
Aquaporinas , Membrana Eritrocítica , Eritrócitos , Peróxido de Hidrogênio , Aquaporinas/metabolismo , Permeabilidade da Membrana Celular , Membrana Eritrocítica/metabolismo , Eritrócitos/metabolismo , Humanos , Peróxido de Hidrogênio/sangue , Peróxido de Hidrogênio/farmacocinética , Metabolismo dos Lipídeos
5.
Kidney Int Rep ; 5(3): 348-357, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32154456

RESUMO

INTRODUCTION: Anion exchanger 1 (AE1) (SLC4A1 gene product) is a membrane protein expressed in both kidney and red blood cells (RBCs): it exchanges extracellular bicarbonate (HCO3 -) for intracellular chloride (Cl-) and participates in acid-base homeostasis. AE1 mutations in kidney α-intercalated cells can lead to distal renal tubular acidosis (dRTA). In RBC, AE1 (known as band 3) is also implicated in membrane stability: deletions can cause South Asian ovalocytosis (SAO). METHODS: We retrospectively collected clinical and biological data from patients harboring dRTA due to a SLC4A1 mutation and analyzed HCO3 - and Cl- transports (by stopped-flow spectrophotometry) and expression (by flow cytometry, fluorescence activated cell sorting, and Coomassie blue staining) in RBCs, as well as RBC membrane stability (ektacytometry). RESULTS: Fifteen patients were included. All experience nephrolithiasis and/or nephrocalcinosis, 2 had SAO and dRTA (dRTA SAO+), 13 dominant dRTA (dRTA SAO-). The latter did not exert specific RBC membrane anomalies. Both HCO3 - and Cl- transports were lower in patients with dRTA SAO+ than in those with dRTA SAO- or controls. Using 3 different extracellular probes, we report a decreased expression (by 52%, P < 0.05) in dRTA SAO+ patients by fluorescence activated cell sorting, whereas total amount of protein was not affected. CONCLUSION: Band 3 transport function and expression in RBCs from dRTA SAO- patients is normal. However, in SAO RBCs, impaired conformation of AE1/band 3 corresponds to an impaired function. Thus, the driver of acid-base defect during dominant dRTA is probably an impaired membrane expression.

6.
Biochim Biophys Acta Biomembr ; 1862(2): 183126, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31738902

RESUMO

Over the past few decades, studies on the red blood cell (RBC) membrane gave rise to increasingly sophisticated although divergent models of its structural organization, since investigations were often performed in denaturing conditions using detergents. To access soluble isolated RBC membrane complexes with the preservation of their interactions and conformations, we decided to apply the recent SMALP (Styrene Maleic Acid Lipid Particles) technology to RBC ghosts. Depending on the ionic strength of buffers in which ghost membranes were resuspended, the isolated proteins within SMALPs could differ on Coomassie-stained gels, but with few changes when compared to ghost membrane SDS lysates. We subsequently produced SMALPs derived from ghosts from two different blood group phenotypes, RhD-positive and RhD-negative, both types of RBC expressing the RhCE proteins but only RhD-positive cells being able to express the RhD proteins. This allowed the isolation, by size exclusion chromatography (SEC), of soluble fractions containing the Rh complex, including the RhD protein or not, within SMALPs. The use a conformation-dependent anti-RhD antibody in immunoprecipitation studies performed on SEC fractions of SMALPs containing Rh proteins clearly demonstrated that the RhD protein, which was only present in SMALPs prepared from RhD-positive RBC ghosts, has preserved at least one important conformational RhD epitope. This approach opens new perspectives in the field of the erythroid membrane study, such as visualization of RBC membrane complexes in native conditions by cryo-electron microscopy (CryoEM) or immuno-tests with conformation-dependent antibodies against blood group antigens on separated and characterized SMALPs containing RBC membrane proteins.


Assuntos
Membrana Eritrocítica/química , Detergentes/química , Membrana Eritrocítica/imunologia , Humanos , Lipossomos/química , Maleatos/química , Proteínas Recombinantes de Fusão/imunologia , Estirenos/química
7.
Sci Rep ; 8(1): 11384, 2018 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-30061676

RESUMO

We previously demonstrated that the translocase protein TSPO2 together with the voltage-dependent anion channel (VDAC) and adenine nucleotide transporter (ANT) were involved in a membrane transport complex in human red blood cells (RBCs). Because VDAC was proposed as a channel mediating ATP release in RBCs, we used TSPO ligands together with VDAC and ANT inhibitors to test this hypothesis. ATP release was activated by TSPO ligands, and blocked by inhibitors of VDAC and ANT, while it was insensitive to pannexin-1 blockers. TSPO ligand increased extracellular ATP (ATPe) concentration by 24-59% over the basal values, displaying an acute increase in [ATPe] to a maximal value, which remained constant thereafter. ATPe kinetics were compatible with VDAC mediating a fast but transient ATP efflux. ATP release was strongly inhibited by PKC and PKA inhibitors as well as by depleting intracellular cAMP or extracellular Ca2+, suggesting a mechanism involving protein kinases. TSPO ligands favoured VDAC polymerization yielding significantly higher densities of oligomeric bands than in unstimulated cells. Polymerization was partially inhibited by decreasing Ca2+ and cAMP contents. The present results show that TSPO ligands induce polymerization of VDAC, coupled to activation of ATP release by a supramolecular complex involving VDAC, TSPO2 and ANT.


Assuntos
Trifosfato de Adenosina/metabolismo , Conexinas/metabolismo , Eritrócitos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Multimerização Proteica , Canais de Ânion Dependentes de Voltagem/metabolismo , Translocador 1 do Nucleotídeo Adenina/metabolismo , Cálcio/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Humanos , Cinética , Ligantes , Modelos Biológicos , Polimerização , Proteína Quinase C/metabolismo , Receptores de GABA/metabolismo
8.
Kidney Int ; 93(2): 390-402, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29054531

RESUMO

Ammonium, stemming from renal ammoniagenesis, is a major urinary proton buffer and is excreted along the collecting duct. This process depends on the concomitant secretion of ammonia by the ammonia channel RhCG and of protons by the vacuolar-type proton-ATPase pump. Thus, urinary ammonium content and urinary acidification are tightly linked. However, mice lacking Rhcg excrete more alkaline urine despite lower urinary ammonium, suggesting an unexpected role of Rhcg in urinary acidification. RhCG and the B1 and B2 proton-ATPase subunits could be co-immunoprecipitated from kidney. In ex vivo microperfused cortical collecting ducts (CCD) proton-ATPase activity was drastically reduced in the absence of Rhcg. Conversely, overexpression of RhCG in HEK293 cells resulted in higher proton secretion rates and increased B1 proton-ATPase mRNA expression. However, in kidneys from Rhcg-/- mice the expression of only B1 and B2 subunits was altered. Immunolocalization of proton-ATPase subunits together with immuno-gold detection of the A proton-ATPase subunit showed similar localization and density of staining in kidneys from Rhcg+/+ and Rhcg-/-mice. In order to test for a reciprocal effect of intercalated cell proton-ATPases on Rhcg activity, we assessed Rhcg and proton-ATPase activities in microperfused CCD from Atp6v1b1-/- mice and showed reduced proton-ATPase activity without altering Rhcg activity. Thus, RhCG and proton-ATPase are located within the same cellular protein complex. RhCG may modulate proton-ATPase function and urinary acidification, whereas proton-ATPase activity does not affect RhCG function. This mechanism may help to coordinate ammonia and proton secretion beyond physicochemical driving forces.


Assuntos
Amônia/urina , Proteínas de Transporte de Cátions/metabolismo , Túbulos Renais Coletores/enzimologia , Glicoproteínas de Membrana/metabolismo , Eliminação Renal , Urina/química , ATPases Vacuolares Próton-Translocadoras/metabolismo , Animais , Proteínas de Transporte de Cátions/deficiência , Proteínas de Transporte de Cátions/genética , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/ultraestrutura , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos Knockout , Complexos Multiproteicos , Prótons , ATPases Vacuolares Próton-Translocadoras/deficiência , ATPases Vacuolares Próton-Translocadoras/genética
9.
Sci Rep ; 7: 46170, 2017 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-28387307

RESUMO

Anion Exchanger 1 (AE1) and stomatin are integral proteins of the red blood cell (RBC) membrane. Erythroid and kidney AE1 play a major role in HCO3- and Cl- exchange. Stomatins down-regulate the activity of many channels and transporters. Biochemical studies suggested an interaction of erythroid AE1 with stomatin. Moreover, we previously reported normal AE1 expression level in stomatin-deficient RBCs. Here, the ability of stomatin to modulate AE1-dependent Cl-/HCO3- exchange was evaluated using stopped-flow methods. In HEK293 cells expressing recombinant AE1 and stomatin, the permeabilities associated with AE1 activity were 30% higher in cells overexpressing stomatin, compared to cells with only endogenous stomatin expression. Ghosts from stomatin-deficient RBCs and controls were resealed in the presence of pH- or chloride-sensitive fluorescent probes and submitted to inward HCO3- and outward Cl- gradients. From alkalinization rate constants, we deduced a 47% decreased permeability to HCO3- for stomatin-deficient patients. Similarly, kinetics of Cl- efflux, followed by the probe dequenching, revealed a significant 42% decrease in patients. In situ Proximity Ligation Assays confirmed an interaction of AE1 with stomatin, in both HEK recombinant cells and RBCs. Here we show that stomatin modulates the transport activity of AE1 through a direct protein-protein interaction.


Assuntos
Proteína 1 de Troca de Ânion do Eritrócito/metabolismo , Proteínas de Membrana/metabolismo , Bicarbonatos/metabolismo , Linhagem Celular , Cloretos/metabolismo , Eritrócitos/metabolismo , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Proteínas de Membrana/genética , Modelos Biológicos , Ligação Proteica
10.
J Am Soc Nephrol ; 28(5): 1507-1520, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27932475

RESUMO

Distal nephron acid secretion is mediated by highly specialized type A intercalated cells (A-ICs), which contain vacuolar H+-ATPase (V-type ATPase)-rich vesicles that fuse with the apical plasma membrane on demand. Intracellular bicarbonate generated by luminal H+ secretion is removed by the basolateral anion-exchanger AE1. Chronically reduced renal acid excretion in distal renal tubular acidosis (dRTA) may lead to nephrocalcinosis and renal failure. Studies in MDCK monolayers led to the proposal of a dominant-negative trafficking mechanism to explain AE1-associated dominant dRTA. To test this hypothesis in vivo, we generated an Ae1 R607H knockin mouse, which corresponds to the most common dominant dRTA mutation in human AE1, R589H. Compared with wild-type mice, heterozygous and homozygous R607H knockin mice displayed incomplete dRTA characterized by compensatory upregulation of the Na+/HCO3- cotransporter NBCn1. Red blood cell Ae1-mediated anion-exchange activity and surface polypeptide expression did not change. Mutant mice expressed far less Ae1 in A-ICs, but basolateral targeting of the mutant protein was preserved. Notably, mutant mice also exhibited reduced expression of V-type ATPase and compromised targeting of this proton pump to the plasma membrane upon acid challenge. Accumulation of p62- and ubiquitin-positive material in A-ICs of knockin mice suggested a defect in the degradative pathway, which may explain the observed loss of A-ICs. R607H knockin did not affect type B intercalated cells. We propose that reduced basolateral anion-exchange activity in A-ICs inhibits trafficking and regulation of V-type ATPase, compromising luminal H+ secretion and possibly lysosomal acidification.


Assuntos
Acidose Tubular Renal/enzimologia , Proteína 1 de Troca de Ânion do Eritrócito/fisiologia , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/enzimologia , ATPases Vacuolares Próton-Translocadoras/fisiologia , Animais , Proteína 1 de Troca de Ânion do Eritrócito/genética , Masculino , Camundongos , Modelos Biológicos
11.
PLoS One ; 8(12): e82338, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24376529

RESUMO

Urea transporter B (UT-B) is a passive membrane channel that facilitates highly efficient permeation of urea. In red blood cells (RBC), while the major function of UT-B is to transport urea, it is assumed that this protein is able to conduct water. Here, we have revisited this last issue by studying RBCs and ghosts from human variants with defects of aquaporin 1 (AQP1) or UT-B. We found that UT-B's osmotic water unit permeability (pfunit) is similar to that of AQP1. The determination of diffusional permeability coefficient (Pd) allowed the calculation of the Pf/Pd ratio, which is consistent with a single-file water transport. Molecular dynamic simulations of water conduction through human UT-B confirmed the experimental finding. From these results, we propose an atomistic description of water-protein interactions involved in this permeation. Inside the UT-B pore, five water molecules were found to form a single-file and move rapidly along a channel by hydrogen bond exchange involving two critical threonines. We further show that the energy barrier for water located in the central region coincides with a water dipole reorientation, which can be related to the proton exclusion observed experimentally. In conclusion, our results indicate that UT-B should be considered as a new member of the water channel family.


Assuntos
Permeabilidade da Membrana Celular , Metabolismo Energético , Eritrócitos/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Água/metabolismo , Sequência de Aminoácidos , Amônia/metabolismo , Animais , Aquaporina 1/metabolismo , Bovinos , Difusão , Membrana Eritrocítica/metabolismo , Humanos , Ligação de Hidrogênio , Cinética , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Osmose , Porosidade , Prótons , Alinhamento de Sequência , Eletricidade Estática , Transportadores de Ureia
12.
PLoS One ; 8(11): e80460, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24260394

RESUMO

Anti-RhD prophylaxis of haemolytic disease of the fetus and newborn (HDFN) is highly effective, but as the suppressive mechanism remains uncertain, a mouse model would be of interest. Here we have generated transgenic mice expressing human RhAG and RhD erythrocyte membrane proteins in the presence and, for human RhAG, in the absence, of mouse Rhag. Human RhAG associates with mouse Rh but not mouse Rhag on red blood cells. In Rhag knockout mice transgenic for human RHAG, the mouse Rh protein is "rescued" (re-expressed), and co-immunoprecipitates with human RhAG, indicating the presence of hetero-complexes which associate mouse and human proteins. RhD antigen was expressed from a human RHD gene on a BAC or from RHD cDNA under control of ß-globin regulatory elements. RhD was never observed alone, strongly indicative that its expression absolutely depends on the presence of transgenic human RhAG. This first expression of RhD in mice is an important step in the creation of a mouse model of RhD allo-immunisation and HDFN, in conjunction with the Rh-Rhag knockout mice we have developed previously.


Assuntos
Proteínas Sanguíneas/genética , Expressão Gênica , Glicoproteínas de Membrana/genética , Sistema do Grupo Sanguíneo Rh-Hr/genética , Compostos de Amônio/metabolismo , Animais , Proteínas Sanguíneas/química , Proteínas Sanguíneas/metabolismo , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Linhagem Celular , Membrana Eritrocítica/metabolismo , Eritrócitos/metabolismo , Regulação da Expressão Gênica , Humanos , Masculino , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Regiões Promotoras Genéticas , Ligação Proteica , Multimerização Proteica , Sistema do Grupo Sanguíneo Rh-Hr/química , Sistema do Grupo Sanguíneo Rh-Hr/metabolismo , Transcrição Gênica , Globinas beta/metabolismo
13.
Am J Physiol Cell Physiol ; 305(6): C654-62, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23842529

RESUMO

Anion exchanger 1 (AE1) or band 3 is a membrane protein responsible for the rapid exchange of chloride for bicarbonate across the red blood cell membrane. Nine mutations leading to single amino-acid substitutions in the transmembrane domain of AE1 are associated with dominant hereditary stomatocytosis, monovalent cation leaks, and reduced anion exchange activity. We set up a stopped-flow spectrofluorometry assay coupled with flow cytometry to investigate the anion transport and membrane expression characteristics of wild-type recombinant AE1 in HEK293 cells, using an inducible expression system. Likewise, study of three stomatocytosis-associated mutations (R730C, E758K, and G796R), allowed the validation of our method. Measurement of the rapid and specific chloride/bicarbonate exchange by surface expressed AE1 showed that E758K mutant was fully active compared with wild-type (WT) AE1, whereas R730C and G796R mutants were inactive, reinforcing previously reported data on other experimental models. Stopped-flow analysis of AE1 transport activity in red blood cell ghost preparations revealed a 50% reduction of G796R compared with WT AE1 corresponding to a loss of function of the G796R mutated protein, in accordance with the heterozygous status of the AE1 variant patients. In conclusion, stopped-flow led to measurement of rapid transport kinetics using the natural substrate for AE1 and, conjugated with flow cytometry, allowed a reliable correlation of chloride/bicarbonate exchange to surface expression of AE1, both in recombinant cells and ghosts and therefore a fine comparison of function between different stomatocytosis samples. This technical approach thus provides significant improvements in anion exchange analysis in red blood cells.


Assuntos
Anemia Hemolítica Congênita/sangue , Proteína 1 de Troca de Ânion do Eritrócito/metabolismo , Bicarbonatos/metabolismo , Cloretos/metabolismo , Eritrócitos/metabolismo , Substituição de Aminoácidos , Anemia Hemolítica Congênita/genética , Anemia Hemolítica Congênita/patologia , Proteína 1 de Troca de Ânion do Eritrócito/genética , Ânions/metabolismo , Linhagem Celular , Eritrócitos/patologia , Células HEK293 , Heterozigoto , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mutação , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
14.
Am J Physiol Cell Physiol ; 302(2): C419-28, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22012326

RESUMO

In red cells, Rh-associated glycoprotein (RhAG) acts as an ammonia channel, as demonstrated by stopped-flow analysis of ghost intracellular pH (pH(i)) changes. Recently, overhydrated hereditary stomatocytosis (OHSt), a rare dominantly inherited hemolytic anemia, was found to be associated with a mutation (Phe65Ser or Ile61Arg) in RHAG. Ghosts from the erythrocytes of four of the OHSt patients with a Phe65Ser mutation were resealed with a pH-sensitive probe and submitted to ammonium gradients. Alkalinization rate constants, reflecting NH(3) transport through the channel and NH(3) diffusion unmediated by RhAG, were deduced from time courses of fluorescence changes. After subtraction of the constant value found for Rh(null) lacking RhAG, we observed that alkalinization rate constant values decreased ∼50% in OHSt compared with those of controls. Similar RhAG expression levels were found in control and OHSt. Since half of the expressed RhAG in OHSt most probably corresponds to the mutated form of RhAG, as expected from the OHSt heterozygous status, this dramatic decrease can be therefore related to the loss of function of the Phe65Ser-mutated RhAG monomer.


Assuntos
Amônia/metabolismo , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Eritrócitos/citologia , Eritrócitos/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Mutação Puntual , Anemia Hemolítica Congênita/genética , Anemia Hemolítica Congênita/metabolismo , Animais , Proteína 1 de Troca de Ânion do Eritrócito/metabolismo , Aquaporina 1/metabolismo , Proteínas Sanguíneas/química , Membrana Eritrocítica/metabolismo , Temperatura Alta , Humanos , Hiperpotassemia/sangue , Hiperpotassemia/congênito , Glicoproteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo
15.
PLoS One ; 5(1): e8921, 2010 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-20126667

RESUMO

BACKGROUND: Rh glycoproteins (RhAG, RhBG, RhCG) are members of the Amt/Mep/Rh family which facilitate movement of ammonium across plasma membranes. Changes in ammonium transport activity following expression of Rh glycoproteins have been described in different heterologous systems such as yeasts, oocytes and eukaryotic cell lines. However, in these complex systems, a potential contribution of endogenous proteins to this function cannot be excluded. To demonstrate that Rh glycoproteins by themselves transport NH(3), human RhCG was purified to homogeneity and reconstituted into liposomes, giving new insights into its channel functional properties. METHODOLOGY/PRINCIPAL FINDINGS: An HA-tag introduced in the second extracellular loop of RhCG was used to purify to homogeneity the HA-tagged RhCG glycoprotein from detergent-solubilized recombinant HEK293E cells. Electron microscopy analysis of negatively stained purified RhCG-HA revealed, after image processing, homogeneous particles of 9 nm diameter with a trimeric protein structure. Reconstitution was performed with sphingomyelin, phosphatidylcholine and phosphatidic acid lipids in the presence of the C(12)E(8) detergent which was subsequently removed by Biobeads. Control of protein incorporation was carried out by freeze-fracture electron microscopy. Particle density in liposomes was a function of the Lipid/Protein ratio. When compared to empty liposomes, ammonium permeability was increased two and three fold in RhCG-proteoliposomes, depending on the Lipid/Protein ratio (1/300 and 1/150, respectively). This strong NH(3) transport was reversibly inhibited by mercuric and copper salts and exhibited a low Arrhenius activation energy. CONCLUSIONS/SIGNIFICANCE: This study allowed the determination of ammonia permeability per RhCG monomer, showing that the apparent Punit(NH3) (around 1x10(-3) microm(3)xs(-1)) is close to the permeability measured in HEK293E cells expressing a recombinant human RhCG (1.60x10(-3) microm(3)xs(-1)), and in human red blood cells endogenously expressing RhAG (2.18x10(-3) microm(3)xs(-1)). The major finding of this study is that RhCG protein is active as an NH(3) channel and that this function does not require any protein partner.


Assuntos
Amônia/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Lipossomos , Glicoproteínas de Membrana/metabolismo , Sequência de Aminoácidos , Biopolímeros , Linhagem Celular , Eletroforese em Gel de Poliacrilamida , Técnica de Fratura por Congelamento , Humanos , Metilaminas/metabolismo , Microscopia Eletrônica de Transmissão , Dados de Sequência Molecular , Osmose , Proteínas Recombinantes/metabolismo
16.
Am J Physiol Cell Physiol ; 297(3): C537-47, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19553567

RESUMO

Rh glycoproteins are members of the ammonium transporter (Amt)/methylamine permease (Mep)/Rh family facilitating movement of NH(3) across plasma membranes. Homology models constructed on the basis of the experimental structures of Escherichia coli AmtB and Nitrosomonas europaea Rh50 indicated a channel structure for human RhA (RhAG), RhB (RhBG), and RhC (RhCG) glycoproteins in which external and internal vestibules are linked by a pore containing two strictly conserved histidines. The pore entry is constricted by two highly conserved phenylalanines, "twin-Phe." In this study, RhCG function was investigated by stopped-flow spectrofluorometry measuring kinetic pH variations in HEK293E cells in the presence of an ammonium gradient. The apparent unitary NH(3) permeability of RhCG was determined and was found to be close to that of AmtB. With a site-directed mutagenesis approach, critical residues involved in Rh NH(3) channel activity were highlighted. In the external vestibule, the importance of both the charge and the conformation of the conserved aspartic acid was shown. In contrast to AmtB, individual mutations of each phenylalanine of the twin-Phe impaired the function while the removal of both resulted in recovery of the transport activity. The impact of the mutations suggests that, although having a common function and a similar channel structure, bacterial AmtB and human Rh vary in several aspects of the NH(3) transport mechanisms.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Glicoproteínas de Membrana/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Proteínas de Transporte de Cátions/genética , Linhagem Celular , Proteínas de Escherichia coli/genética , Humanos , Concentração de Íons de Hidrogênio , Glicoproteínas de Membrana/genética , Modelos Moleculares , Mutação , Conformação Proteica , Compostos de Amônio Quaternário/química , Compostos de Amônio Quaternário/metabolismo , Alinhamento de Sequência , Espectrometria de Fluorescência
17.
Biochem J ; 391(Pt 1): 33-40, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15929723

RESUMO

The mammalian Rh (Rhesus) protein family belongs to the Amt/Mep (ammonia transporter/methylammonium permease)/Rh superfamily of ammonium transporters. Whereas RhCE, RhD and RhAG are erythroid specific, RhBG and RhCG are expressed in key organs associated with ammonium transport and metabolism. We have investigated the ammonium transport function of human RhBG and RhCG by comparing intracellular pH variation in wild-type and transfected HEK-293 (human embryonic kidney) cells and MDCK (Madin-Darby canine kidney) cells in the presence of ammonium (NH4+/NH3) gradients. Stopped-flow spectrofluorimetry analysis, using BCECF [2',7'-bis-(2-carboxyethyl)-5(6)-carboxyfluorescein] as a pH-sensitive probe, revealed that all cells submitted to inwardly or outwardly directed ammonium gradients exhibited rapid alkalinization or acidification phases respectively, which account for ammonium movements in transfected and native cells. However, as compared with wild-type cells known to have high NH3 lipid permeability, RhBG- and RhCG-expressing cells exhibited ammonium transport characterized by: (i) a five to six times greater kinetic rate-constant; (ii) a weak temperature-dependence; and (iii) reversible inhibition by mercuric chloride (IC50: 52 microM). Similarly, when subjected to a methylammonium gradient, RhBG- and RhCG-expressing cells exhibited kinetic rate constants greater than those of native cells. However, these constants were five times higher for RhBG as compared with RhCG, suggesting a difference in substrate accessibility. These results, indicating that RhBG and RhCG facilitate rapid and low-energy-dependent bi-directional ammonium movement across the plasma membrane, favour the hypothesis that these Rh glycoproteins, together with their erythroid homologue RhAG [Ripoche, Bertrand, Gane, Birkenmeier, Colin and Cartron (2005) Proc. Natl. Acad. Sci. U.S.A. 101, 17222-17227] constitute a family of NH3 channels in mammalian cells.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Glicoproteínas/metabolismo , Rim/citologia , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Compostos de Amônio Quaternário/metabolismo , Animais , Transporte Biológico , Proteínas de Transporte de Cátions/genética , Linhagem Celular , Membrana Celular/metabolismo , Permeabilidade da Membrana Celular/efeitos dos fármacos , Cães , Citometria de Fluxo , Glicoproteínas/genética , Humanos , Concentração de Íons de Hidrogênio , Cinética , Glicoproteínas de Membrana/genética , Proteínas de Membrana Transportadoras/genética , Cloreto de Mercúrio/farmacologia , Metilaminas/metabolismo , Mutagênese Sítio-Dirigida , Especificidade por Substrato
18.
J Biol Chem ; 278(28): 25526-33, 2003 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-12719424

RESUMO

Several studies suggest that the Rh complex represents a major interaction site between the membrane lipid bilayer and the red cell skeleton, but little is known about the molecular basis of this interaction. We report here that ankyrin-R is capable of interacting directly with the C-terminal cytoplasmic domain of Rh and RhAG polypeptides. We first show that the primary defect of ankyrin-R in normoblastosis (nb/nb) spherocytosis mutant mice is associated with a sharp reduction of RhAG and Rh polypeptides. Secondly, our flow cytometric analysis of the Triton X-100 extractability of recombinant fusion proteins expressed in erythroleukemic cell lines suggests that the C-terminal cytoplasmic domains of Rh and RhAG are sufficient to mediate interaction with the erythroid membrane skeleton. Using the yeast two-hybrid system, we demonstrate a direct interaction between the cytoplasmic tails of Rh and RhAG and the second repeat domain (D2) of ankyrin-R. This finding is supported by the demonstration that the substitution of Asp-399 in the cytoplasmic tail of RhAG, a mutation associated with the deficiency of the Rh complex in one Rhnull patient, totally impaired interaction with domain D2 of ankyrin-R. These results identify the Rh/RhAG-ankyrin complex as a new interaction site between the red cell membrane and the spectrin-based skeleton, the disruption of which might result in the stomato-spherocytosis typical of Rhnull red cells.


Assuntos
Anquirinas/química , Anquirinas/metabolismo , Proteínas Sanguíneas , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Animais , Western Blotting , Proteínas de Ligação a Calmodulina/metabolismo , Citoplasma/metabolismo , Detergentes/farmacologia , Eletroforese em Gel de Poliacrilamida , Eritrócitos/metabolismo , Citometria de Fluxo , Glutationa Transferase/metabolismo , Humanos , Células K562 , Bicamadas Lipídicas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Mutantes , Modelos Biológicos , Octoxinol/farmacologia , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Sistema do Grupo Sanguíneo Rh-Hr/química , Transfecção , Técnicas do Sistema de Duplo-Híbrido
19.
Blood ; 101(1): 338-44, 2003 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12393467

RESUMO

Rh(null) red cells are characteristically stomato-spherocytic. This and other evidence suggest that the Rh complex represents a major attachment site between the membrane lipid bilayer and the erythroid skeleton. As an attempt to identify the linking protein(s) between the red cell skeleton and the Rh complex, we analyzed the expression of Rh, RhAG, CD47, LW, and glycophorin B proteins in red cells from patients with hereditary spherocytosis associated with complete protein 4.2 deficiency but normal band 3 (4.2(-)HS). Flow cytometric and immunoblotting analysis revealed a severe reduction of CD47 (up to 80%) and a slower mobility of RhAG on sodium dodecyl sulfate-polyacrylamide gel electrophoresis, possibly reflecting an overglycosylation state. Unexpectedly, 4.2(-/-) mice, which are anemic, displayed a normal red cell expression of CD47 and RhAG. These results suggest that human protein 4.2, through interaction with CD47, is involved in the skeleton linkage and/or membrane translocation of the Rh complex. However, these potential role(s) of protein 4.2 might be not conserved across species. Finally, the absence or low expression of red cell CD47 in CD47(-/-) mice and in some humans carrying RHCE gene variants (D--, D., and R(N)), respectively, had no detectable effect on protein 4.2 and RhAG expression. Since these cells are morphologically normal with no sign of hemolysis, it is assumed that CD47 deficiency per se is not responsible for the cell shape abnormalities and for the compensated hemolytic anemia typical of 4.2(-) and Rh(null) red cells.


Assuntos
Antígenos CD/metabolismo , Proteínas Sanguíneas/metabolismo , Proteínas de Transporte/metabolismo , Animais , Proteínas Sanguíneas/genética , Antígeno CD47 , Proteínas do Citoesqueleto , Membrana Eritrocítica/metabolismo , Humanos , Glicoproteínas de Membrana , Proteínas de Membrana , Camundongos , Camundongos Knockout , Mutação , Ligação Proteica , Sistema do Grupo Sanguíneo Rh-Hr , Esferocitose Hereditária/genética , Esferocitose Hereditária/metabolismo
20.
Blood ; 100(3): 1038-47, 2002 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12130520

RESUMO

In most cases, the lack of Rh in Rh(null) red cells is associated with RHAG gene mutations. We explored the role of RhAG in the surface expression of Rh. Nonerythroid HEK293 cells, which lack Rh and RhAG, or erythroid K562 cells, which endogenously express RhAG but not Rh, were transfected with RhD and/or RhAG cDNAs using cytomegalovirus (CMV) promoter-based expression vectors. In HEK293 cells, a low but significant expression of RhD was obtained only when RhAG was expressed at a high level. In K562 cells, as expected from the opposite effects of the phorbol ester 12-O-tetradecanoyl phorbol 13-acetate (TPA) on erythroid and CMV promoters, the levels of endogenous RhAG and recombinant RhD transcripts were substantially decreased and enhanced upon TPA treatment of RhD-transfected cells (K562/RhD), respectively. However, flow cytometry and fluorescence microscopy analysis revealed a decreased cell-surface expression of both RhAG and RhD proteins. Conversely, TPA treatment of RhAG-transfected cells increased both the transcript and surface expression levels of RhAG. When K562/RhD cells were cotransfected by the RhAG cDNA, the TPA-mediated induction of recombinant RhAG and RhD transcription was associated with an increased membrane expression of both RhAG and RhD proteins. These results demonstrate the role of RhAG as a strictly required posttranscriptional factor regulating Rh membrane expression. In addition, because the postulated 2:2 stoichiometry between Rh and RhAG observed in the native red cell membrane could not be obtained in cotransfected K562 cells, our study also suggests that as yet unidentified protein(s) might be involved for optimal membrane expression of Rh.


Assuntos
Antígenos de Superfície/efeitos dos fármacos , Proteínas Sanguíneas , Glicoproteínas de Membrana/fisiologia , Sistema do Grupo Sanguíneo Rh-Hr/metabolismo , Antígenos de Superfície/química , Antígenos de Superfície/metabolismo , Citometria de Fluxo , Regulação da Expressão Gênica , Humanos , Células K562 , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Microscopia de Fluorescência , Transporte Proteico/efeitos dos fármacos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sistema do Grupo Sanguíneo Rh-Hr/química , Acetato de Tetradecanoilforbol/farmacologia , Transcrição Gênica/efeitos dos fármacos , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA