Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Cell Sci ; 129(6): 1179-89, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26823605

RESUMEN

The cellular machinery responsible for Cu(+)-stimulated delivery of the Wilson-disease-associated protein ATP7B to the apical domain of hepatocytes is poorly understood. We demonstrate that myosin Vb regulates the Cu(+)-stimulated delivery of ATP7B to the apical domain of polarized hepatic cells, and that disruption of the ATP7B-myosin Vb interaction reduces the apical surface expression of ATP7B. Overexpression of the myosin Vb tail, which competes for binding of subapical cargos to myosin Vb bound to subapical actin, disrupted the surface expression of ATP7B, leading to reduced cellular Cu(+) export. The myosin-Vb-dependent targeting step occurred in parallel with hepatocyte-like polarity. If the myosin Vb tail was expressed acutely in cells just prior to the establishment of polarity, it appeared as part of an intracellular apical compartment, centered on γ-tubulin. ATP7B became selectively arrested in this compartment at high [Cu(+)] in the presence of myosin Vb tail, suggesting that these compartments are precursors of donor-acceptor transfer stations for apically targeted cargos of myosin Vb. Our data suggest that reduced hepatic Cu(+) clearance in idiopathic non-Wilsonian types of disease might be associated with the loss of function of myosin Vb.


Asunto(s)
Polaridad Celular , Cobre/metabolismo , Hepatocitos/metabolismo , Degeneración Hepatolenticular/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo V/metabolismo , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Línea Celular , ATPasas Transportadoras de Cobre , Hepatocitos/citología , Degeneración Hepatolenticular/genética , Humanos , Hígado/citología , Hígado/metabolismo , Cadenas Pesadas de Miosina/genética , Miosina Tipo V/genética , Transporte de Proteínas
2.
Proc Natl Acad Sci U S A ; 111(14): E1364-73, 2014 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-24706876

RESUMEN

Wilson disease (WD) is a monogenic autosomal-recessive disorder of copper accumulation that leads to liver failure and/or neurological deficits. WD is caused by mutations in ATP7B, a transporter that loads Cu(I) onto newly synthesized cupro-enzymes in the trans-Golgi network (TGN) and exports excess copper out of cells by trafficking from the TGN to the plasma membrane. To date, most WD mutations have been shown to disrupt ATP7B activity and/or stability. Using a multidisciplinary approach, including clinical analysis of patients, cell-based assays, and computational studies, we characterized a patient mutation, ATP7B(S653Y), which is stable, does not disrupt Cu(I) transport, yet renders the protein unable to exit the TGN. Bulky or charged substitutions at position 653 mimic the phenotype of the patient mutation. Molecular modeling and dynamic simulation suggest that the S653Y mutation induces local distortions within the transmembrane (TM) domain 1 and alter TM1 interaction with TM2. S653Y abolishes the trafficking-stimulating effects of a secondary mutation in the N-terminal apical targeting domain. This result indicates a role for TM1/TM2 in regulating conformations of cytosolic domains involved in ATP7B trafficking. Taken together, our experiments revealed an unexpected role for TM1/TM2 in copper-regulated trafficking of ATP7B and defined a unique class of WD mutants that are transport-competent but trafficking-defective. Understanding the precise consequences of WD-causing mutations will facilitate the development of advanced mutation-specific therapies.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Transporte de Catión/metabolismo , Cobre/metabolismo , Degeneración Hepatolenticular/genética , Mutación , Adenosina Trifosfatasas/química , Secuencia de Aminoácidos , Animales , Proteínas de Transporte de Catión/química , Membrana Celular/metabolismo , ATPasas Transportadoras de Cobre , Aparato de Golgi/metabolismo , Humanos , Hígado/metabolismo , Modelos Moleculares , Modelos Teóricos , Datos de Secuencia Molecular , Transporte de Proteínas , Homología de Secuencia de Aminoácido
3.
Traffic ; 15(12): 1344-65, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25243755

RESUMEN

Physiologic Cu levels regulate the intracellular location of the Cu ATPase ATP7B. Here, we determined the routes of Cu-directed trafficking of endogenous ATP7B in the polarized hepatic cell line WIF-B and in the liver in vivo. Copper (10 µm) caused ATP7B to exit the trans-Golgi network (TGN) in vesicles, which trafficked via large basolateral endosomes to the apical domain within 1 h. Although perturbants of luminal acidification had little effect on the TGN localization of ATP7B in low Cu, they blocked delivery to the apical membrane in elevated Cu. If the vesicular proton-pump inhibitor bafilomycin-A1 (Baf) was present with Cu, ATP7B still exited the TGN, but accumulated in large endosomes located near the coverslip, in the basolateral region. Baf washout restored ATP7B trafficking to the apical domain. If ATP7B was staged apically in high Cu, Baf addition promoted the accumulation of ATP7B in subapical endosomes, indicating a blockade of apical recycling, with concomitant loss of ATP7B at the apical membrane. The retrograde pathway to the TGN, induced by Cu removal, was far less affected by Baf than the anterograde (Cu-stimulated) case. Overall, loss of acidification-impaired Cu-regulated trafficking of ATP7B at two main sites: (i) sorting and exit from large basolateral endosomes and (ii) recycling via endosomes near the apical membrane.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Transporte de Catión/metabolismo , Cobre/metabolismo , Endosomas/metabolismo , Hepatocitos/metabolismo , Animales , Línea Celular Tumoral , ATPasas Transportadoras de Cobre , Hepatocitos/efectos de los fármacos , Macrólidos/farmacología , Transporte de Proteínas , Ratas , Red trans-Golgi/metabolismo
4.
J Biol Chem ; 290(14): 8803-19, 2015 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-25666620

RESUMEN

The Wilson disease protein ATP7B exhibits copper-dependent trafficking. In high copper, ATP7B exits the trans-Golgi network and moves to the apical domain of hepatocytes where it facilitates elimination of excess copper through the bile. Copper levels also affect ATP7B phosphorylation. ATP7B is basally phosphorylated in low copper and becomes more phosphorylated ("hyperphosphorylated") in elevated copper. The functional significance of hyperphosphorylation remains unclear. We showed that hyperphosphorylation occurs even when ATP7B is restricted to the trans-Golgi network. We performed comprehensive phosphoproteomics of ATP7B in low versus high copper, which revealed that 24 Ser/Thr residues in ATP7B could be phosphorylated, and only four of these were copper-responsive. Most of the phosphorylated sites were found in the N- and C-terminal cytoplasmic domains. Using truncation and mutagenesis, we showed that inactivation or elimination of all six N-terminal metal binding domains did not block copper-dependent, reversible, apical trafficking but did block hyperphosphorylation in hepatic cells. We showed that nine of 15 Ser/Thr residues in the C-terminal domain were phosphorylated. Inactivation of 13 C-terminal phosphorylation sites reduced basal phosphorylation and eliminated hyperphosphorylation, suggesting that copper binding at the N terminus propagates to the ATP7B C-terminal region. C-terminal mutants with either inactivating or phosphomimetic substitutions showed little effect upon copper-stimulated trafficking, indicating that trafficking does not depend on phosphorylation at these sites. Thus, our studies revealed that copper-dependent conformational changes in the N-terminal region lead to hyperphosphorylation at C-terminal sites, which seem not to affect trafficking and may instead fine-tune copper sequestration.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Transporte de Catión/metabolismo , Cobre/metabolismo , Serina/metabolismo , Treonina/metabolismo , Adenosina Trifosfatasas/química , Secuencia de Aminoácidos , Animales , Transporte Biológico , Proteínas de Transporte de Catión/química , Línea Celular , ATPasas Transportadoras de Cobre , Humanos , Datos de Secuencia Molecular , Fosforilación
5.
J Cell Sci ; 127(Pt 16): 3535-45, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24928903

RESUMEN

The intestinal brush border Na(+)/H(+) exchanger NHE3 is tightly regulated through changes in its endocytosis and exocytosis. Myosin VI, a minus-end-directed actin motor, has been implicated in endocytosis at the inter-microvillar cleft and during vesicle remodeling in the terminal web. Here, we asked whether myosin VI also regulates NHE3 movement down the microvillus. The basal NHE3 activity and its surface amount, determined by fluorometry of the ratiometric pH indicator BCECF and biotinylation assays, respectively, were increased in myosin-VI-knockdown (KD) Caco-2/Bbe cells. Carbachol (CCH) and forskolin (FSK) stimulated NHE3 endocytosis in control but not in myosin VI KD cells. Importantly, immunoelectron microscopy results showed that NHE3 was preferentially localized in the basal half of control microvilli but in the distal half in myosin VI KD cells. Treatment with dynasore duplicated some aspects of myosin VI KD: it increased basal surface NHE3 activity and prevented FSK-induced NHE3 endocytosis. However, NHE3 had an intermediate distribution along the microvillus (between that in myosin VI KD and untreated cells) in dynasore-treated cells. We conclude that myosin VI is required for basal and stimulated endocytosis of NHE3 in intestinal cells, and suggest that myosin VI also moves NHE3 down the microvillus.


Asunto(s)
Células Epiteliales/metabolismo , Intestinos/citología , Microvellosidades/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Animales , Carbacol/metabolismo , Línea Celular , Endocitosis , Humanos , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microvellosidades/genética , Cadenas Pesadas de Miosina/genética , Transporte de Proteínas , Intercambiador 3 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/genética
6.
Proc Natl Acad Sci U S A ; 108(13): 5390-5, 2011 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-21406592

RESUMEN

In human disorders, the genotype-phenotype relationships are often complex and influenced by genetic and/or environmental factors. Wilson disease (WD) is a monogenic disorder caused by mutations in the copper-transporting P-type ATPase ATP7B. WD shows significant phenotypic diversity even in patients carrying identical mutations; the basis for such diverse manifestations is unknown. We demonstrate that the 2623A/G polymorphism (producing the Gly(875) → Arg substitution in the A-domain of ATP7B) drastically alters the intracellular properties of ATP7B, whereas copper reverses the effects. Under basal conditions, the common Gly(875) variant of ATP7B is targeted to the trans-Golgi network (TGN) and transports copper into the TGN lumen. In contrast, the Arg(875) variant is located in the endoplasmic reticulum (ER) and does not deliver copper to the TGN. Elevated copper corrects the ATP7B-Arg(875) phenotype. Addition of only 0.5-5 µM copper triggers the exit of ATP7B-Arg(875) from the ER and restores copper delivery to the TGN. Analysis of the recombinant A-domains by NMR suggests that the ER retention of ATP7B-Arg(875) is attributable to increased unfolding of the Arg(875)-containing A-domain. Copper is not required for the folding of ATP7B-Arg(875) during biosynthesis, but it stabilizes protein and stimulates its activity. A chemotherapeutical drug, cisplatin, that mimics a copper-bound state of ATP7B also corrects the "disease-like" phenotype of ATP7B-Arg(875) and promotes its TGN targeting and transport function. We conclude that in populations harboring the Arg(875) polymorphism, the levels of bioavailable copper may play a vital role in the manifestations of WD.


Asunto(s)
Adenosina Trifosfatasas/genética , Arginina/genética , Proteínas de Transporte de Catión/genética , Cobre/metabolismo , Fenotipo , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/metabolismo , Proteínas de Transporte de Catión/química , Proteínas de Transporte de Catión/metabolismo , Línea Celular , ATPasas Transportadoras de Cobre , Retículo Endoplásmico/metabolismo , Humanos , Modelos Moleculares , Polimorfismo Genético , Conformación Proteica , Red trans-Golgi/metabolismo
7.
J Biol Chem ; 286(26): 22833-45, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21561868

RESUMEN

ClC-5, a chloride/proton exchanger, is predominantly expressed and localized in subapical endosomes of the renal proximal tubule. Mutations of the CLCN5 gene cause Dent disease. The symptoms of Dent disease are replicated in Clcn5 knock-out mice. Absence of ClC-5 in mice is associated with reduced surface expression of NHE3 in proximal tubules. The molecular basis for this change is not fully understood. In this study, we investigated the mechanisms by which ClC-5 regulates trafficking of NHE3. Whether ClC-5-dependent endocytosis, exocytosis, or both contributed to the altered distribution of NHE3 was examined. First, NHE3 activity in proximal tubules of wild type (WT) and Clcn5 KO mice was determined by two-photon microscopy. Basal and dexamethasone-stimulated NHE3 activity of Clcn5 KO mice was decreased compared with that seen in WT mice, whereas the degree of inhibition of NHE3 activity by increasing cellular concentration of cAMP (forskolin) or Ca(2+) (A23187) was not different in WT and Clcn5 KO mice. Second, NHE3-dependent absorption of HCO(3)(-), measured by single tubule perfusion, was reduced in proximal tubules of Clcn5 KO mice. Third, by cell surface biotinylation, trafficking of NHE3 was examined in short hairpin RNA (shRNA) plasmid-transfected opossum kidney cells. Surface NHE3 was reduced in opossum kidney cells with reduced expression of ClC-5, whereas the total protein level of NHE3 did not change. Parathyroid hormone decreased NHE3 surface expression, but the extent of decrease and the rate of endocytosis observed in both scrambled and ClC-5 knockdown cells were not significantly different. However, the rates of basal and dexamethasone-stimulated exocytosis of NHE3 were attenuated in ClC-5 knockdown cells. These results show that ClC-5 plays an essential role in exocytosis of NHE3.


Asunto(s)
Canales de Cloruro/metabolismo , Enfermedad de Dent/metabolismo , Exocitosis , Túbulos Renales Proximales/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Animales , Bicarbonatos/metabolismo , Línea Celular , Canales de Cloruro/genética , Enfermedad de Dent/genética , Modelos Animales de Enfermedad , Endocitosis/efectos de los fármacos , Endocitosis/genética , Humanos , Ratones , Ratones Noqueados , Zarigüeyas , Hormona Paratiroidea/farmacología , Transporte de Proteínas/genética , Intercambiador 3 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/genética
8.
Traffic ; 10(6): 767-79, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19416479

RESUMEN

Human Cu-ATPases ATP7A and ATP7B maintain copper homeostasis through regulated trafficking between intracellular compartments. Inactivation of these transporters causes Menkes disease and Wilson disease, respectively. In Menkes disease, copper accumulates in kidneys and causes tubular damage, indicating that the renal ATP7B does not compensate for the loss of ATP7A function. We show that this is likely due to a kidney-specific regulation of ATP7B. Unlike ATP7A (or hepatic ATP7B) which traffics from the TGN to export copper, renal ATP7B does not traffic and therefore is unlikely to mediate copper export. The lack of ATP7B trafficking is not on account of the loss of a kinase-mediated phosphorylation or simultaneous presence of ATP7A in renal cells. Rather, the renal ATP7B appears 2-3 kDa smaller than hepatic ATP7B. Recombinant ATP7B expressed in renal cells is similar to hepatic protein in size and trafficking. The analysis of ATP7B mRNA revealed a complex behavior of exon 1 upon amplification, suggesting that it could be inefficiently translated. Recombinant ATP7B lacking exon 1 traffics differently in renal and hepatic cells, but does not fully recapitulate the endogenous phenotype. We discuss factors that may contribute to cell-specific behavior of ATP7B and propose a role for renal ATP7B in intracellular copper storage.


Asunto(s)
Adenosina Trifosfatasas/fisiología , Proteínas de Transporte de Catión/fisiología , Cobre/metabolismo , Riñón/fisiología , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de Transporte de Catión/química , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Línea Celular , ATPasas Transportadoras de Cobre , Exones , Humanos , Riñón/metabolismo , Datos de Secuencia Molecular , Fosforilación , Transporte de Proteínas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido
9.
Am J Physiol Cell Physiol ; 301(5): C1140-9, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21832249

RESUMEN

Gastrointestinal infection with Shiga toxins producing enterohemorrhagic Escherichia coli causes the spectrum of gastrointestinal and systemic complications, including hemorrhagic colitis and hemolytic uremic syndrome, which is fatal in ∼10% of patients. However, the molecular mechanisms of Stx endocytosis by enterocytes and the toxins cross the intestinal epithelium are largely uncharacterized. We have studied Shiga toxin 1 entry into enterohemorrhagic E. coli-infected intestinal epithelial cells and found that bacteria stimulate Shiga toxin 1 macropinocytosis through actin remodeling. This enterohemorrhagic E. coli-caused macropinocytosis occurs through a nonmuscle myosin II and cell division control 42 (Cdc42)-dependent mechanism. Macropinocytosis of Shiga toxin 1 is followed by its transcytosis to the basolateral environment, a step that is necessary for its systemic spread. Inhibition of Shiga toxin 1 macropinocytosis significantly decreases toxin uptake by intestinal epithelial cells and in this way provides an attractive, antibiotic-independent strategy for prevention of the harmful consequences of enterohemorrhagic E. coli infection.


Asunto(s)
Escherichia coli Enterohemorrágica , Infecciones por Escherichia coli/metabolismo , Mucosa Intestinal/metabolismo , Pinocitosis , Toxina Shiga I/metabolismo , Transcitosis , Actinas/metabolismo , Línea Celular , Colon/metabolismo , Colon/microbiología , Infecciones por Escherichia coli/microbiología , Humanos , Mucosa Intestinal/microbiología , Miosina Tipo II/metabolismo , Proteína de Unión al GTP cdc42/metabolismo
10.
Biometals ; 24(1): 159-70, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20981470

RESUMEN

Manganese is a trace element that is an essential co-factor in many enzymes critical to diverse biological pathways. However, excess Mn(2+) leads to neurotoxicity, with psychiatric and motor dysfunction resembling parkinsonism. The liver is the main organ for Mn(2+) detoxification by excretion into bile. Although many pathways of cellular Mn(2+) uptake have been established, efflux mechanisms remain essentially undefined. In this study, we evaluated a potential role in Mn(2+) detoxification by the Secretory Pathway Ca(2+), Mn(2+)-ATPase in rat liver and a liver-derived cell model WIF-B that polarizes to distinct bile canalicular and sinusoidal domains in culture. Of two known isoforms, only secretory pathway Ca(2+)-ATPase isoform 1 (SPCA1) was expressed in liver and WIF-B cells. As previously observed in non-polarized cells, SPCA1 showed overlapping distribution with TGN38, consistent with Golgi/TGN localization. However, a prominent novel localization of SPCA1 to an endosomal population close to, but not on the basolateral membrane was also observed. This was confirmed by fractionation of rat liver homogenates which revealed dual distribution of SPCA1 to the Golgi/TGN and a fraction that included the early endosomal marker, EEA1. We suggest that this novel pool of endosomes may serve to sequester Mn(2+) as it enters from the sinusoidal/basolateral domains. Isoform-specific partial knockdown of SPCA1 delayed cell growth and formation of canalicular domain by about 30% and diminished viability upon exposure to Mn(2+). Conversely, overexpression of SPCA1 in HEK 293T cells conferred tolerance to Mn(2+) toxicity. Taken together, our findings suggest a role for SPCA1 in Mn(2+) detoxification in liver.


Asunto(s)
ATPasas Transportadoras de Calcio/metabolismo , Hígado/citología , Manganeso/metabolismo , Manganeso/toxicidad , Animales , ATPasas Transportadoras de Calcio/genética , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células HEK293 , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Ratas
11.
Mol Biol Cell ; 17(6): 2661-73, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16540524

RESUMEN

Based on physiological studies, the epithelial brush-border (BB) Na+/H+ antiporter3 (NHE3) seems to associate with the actin cytoskeleton both by binding to and independently of the PDZ domain containing proteins NHERF1 and NHERF2. We now show that NHE3 directly binds ezrin at a site in its C terminus between aa 475-589, which is separate from the PSD95/dlg/zonular occludens-1 (PDZ) interacting domain. This is an area predicted to be alpha-helical, with a positive aa cluster on one side (K516, R520, and R527). Point mutations of these positively charged aa reduced (NHE3 double mutant [R520F, R527F]) or abolished (NHE3 triple mutant [K516Q, R520F, R 527F]) ezrin binding. Functional consequences of these NHE3 point mutants included the following. 1) A marked decrease in surface amount with a greater decrease in NHE3 activity. 2) Decreased surface expression due to decreased rates of exocytosis and plasma membrane delivery of newly synthesized NHE3, with normal total expression levels and slightly reduced endocytosis rates. 3) A longer plasma membrane half-life of mutant NHE3 with normal total half-life. 4) Decreased BB mobile fraction of NHE3 double mutant. These results show that NHE3 binds ezrin directly as well as indirectly and suggest that the former is related to 1) the exocytic trafficking of and plasma membrane delivery of newly synthesized NHE3, which determines the amount of plasma membrane NHE3 and partially determines NHE3 activity, and 2) BB mobility of NHE3, which may increase its delivery from microvilli to the intervillus clefts, perhaps for NHE3-regulated endocytosis.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Aparato Yuxtaglomerular/metabolismo , Intercambiadores de Sodio-Hidrógeno/genética , Intercambiadores de Sodio-Hidrógeno/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Membrana Celular/metabolismo , Secuencia Conservada , Citoplasma/metabolismo , Humanos , Cinética , Datos de Secuencia Molecular , Transporte de Proteínas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Intercambiadores de Sodio-Hidrógeno/química
12.
Mol Biol Cell ; 13(10): 3400-15, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12388745

RESUMEN

Membrane trafficking is central to establishing and maintaining epithelial cell polarity. One open question is to what extent the mechanisms regulating membrane trafficking are conserved between nonpolarized and polarized cells. To answer this question, we examined the dynamics of domain-specific plasma membrane (PM) proteins in three classes of hepatic cells: polarized and differentiated WIF-B cells, nonpolarized and differentiated Fao cells, and nonpolarized and nondifferentiated Clone 9 cells. In nonpolarized cells, mature apical proteins were uniformly distributed in the PM. Surprisingly, they were also in an intracellular compartment. Double labeling revealed that the compartment contained only apical proteins. By monitoring the dynamics of antibody-labeled molecules in nonpolarized cells, we further found that apical proteins rapidly recycled between the compartment and PM. In contrast, the apical PM residents in polarized cells showed neither internalization nor return to the basolateral PM from which they had originally come. Cytochalasin D treatment of these polarized cells revealed that the retention mechanisms are actin dependent. We conclude from these data that both polarized and nonpolarized cells selectively sort apical proteins from the PM and transport them to specific, but different cellular locations. We propose that the intracellular recycling compartment in nonpolarized cells is an intermediate in apical surface formation.


Asunto(s)
Membrana Celular/metabolismo , Polaridad Celular , Hepatocitos/metabolismo , Proteínas de la Membrana/metabolismo , Transporte de Proteínas/fisiología , Actinas/metabolismo , Animales , Biomarcadores , Compartimento Celular , Línea Celular , Hepatocitos/citología , Modelos Biológicos , Estructura Terciaria de Proteína , Ratas
13.
Mol Biol Cell ; 14(7): 2689-705, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12857857

RESUMEN

We examined the role that lipid rafts play in regulating apical protein trafficking in polarized hepatic cells. Rafts are postulated to form in the trans-Golgi network where they recruit newly synthesized apical residents and mediate their direct transport to the apical plasma membrane. In hepatocytes, single transmembrane and glycolipid-anchored apical proteins take the "indirect" route. They are transported from the trans-Golgi to the basolateral plasma membrane where they are endocytosed and transcytosed to the apical surface. Do rafts sort hepatic apical proteins along this circuitous pathway? We took two approaches to answer this question. First, we determined the detergent solubility of selected apical proteins and where in the biosynthetic pathway insolubility was acquired. Second, we used pharmacological agents to deplete raft components and assessed their effects on basolateral-to-apical transcytosis. We found that cholesterol and glycosphingolipids are required for delivery from basolateral early endosomes to the subapical compartment. In contrast, fluid phase uptake and clathrin-mediated internalization of recycling receptors were only mildly impaired. Apical protein solubility did not correlate with raft depletion or impaired transcytosis, suggesting other factors contribute to apical protein insolubility. Examination of apical proteins in Fao cells also revealed that raft-dependent sorting does not require the polarized cell context.


Asunto(s)
Colesterol/metabolismo , Endosomas/metabolismo , Glicoesfingolípidos/metabolismo , Hepatocitos/metabolismo , Animales , Transporte Biológico/fisiología , Membrana Celular/metabolismo , Membrana Celular/fisiología , Polaridad Celular/fisiología , Células Cultivadas , Colesterol/fisiología , Clonación Molecular , Detergentes/farmacología , Endocitosis/fisiología , Endosomas/fisiología , Glicoesfingolípidos/fisiología , Aparato de Golgi/metabolismo , Aparato de Golgi/fisiología , Hepatocitos/fisiología , Microdominios de Membrana/metabolismo , Microdominios de Membrana/fisiología , Ratas , Solubilidad/efectos de los fármacos , Red trans-Golgi/metabolismo , Red trans-Golgi/fisiología
16.
PLoS One ; 8(7): e69196, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23874912

RESUMEN

Life-threatening intestinal and systemic effects of the Shiga toxins produced by enterohemorrhagic Escherichia coli (EHEC) require toxin uptake and transcytosis across intestinal epithelial cells. We have recently demonstrated that EHEC infection of intestinal epithelial cells stimulates toxin macropinocytosis, an actin-dependent endocytic pathway. Host actin rearrangement necessary for EHEC attachment to enterocytes is mediated by the type 3 secretion system which functions as a molecular syringe to translocate bacterial effector proteins directly into host cells. Actin-dependent EHEC attachment also requires the outer membrane protein intimin, a major EHEC adhesin. Here, we investigate the role of type 3 secretion in actin turnover occurring during toxin macropinocytosis. Toxin macropinocytosis is independent of EHEC type 3 secretion and intimin attachment. EHEC soluble factors are sufficient to stimulate macropinocytosis and deliver toxin into enterocytes in vitro and in vivo; intact bacteria are not required. Intimin-negative enteroaggregative Escherichia coli (EAEC) O104:H4 robustly stimulate Shiga toxin macropinocytosis into intestinal epithelial cells. The apical macropinosomes formed in intestinal epithelial cells move through the cells and release their cargo at these cells' basolateral sides. Further analysis of EHEC secreted proteins shows that a serine protease EspP alone is able to stimulate host actin remodeling and toxin macropinocytosis. The observation that soluble factors, possibly serine proteases including EspP, from each of two genetically distinct toxin-producing strains, can stimulate Shiga toxin macropinocytosis and transcellular transcytosis alters current ideas concerning mechanisms whereby Shiga toxin interacts with human enterocytes. Mechanisms important for this macropinocytic pathway could suggest new potential therapeutic targets for Shiga toxin-induced disease.


Asunto(s)
Escherichia coli Enterohemorrágica/enzimología , Proteínas de Escherichia coli/metabolismo , Mucosa Intestinal/metabolismo , Pinocitosis/fisiología , Serina Endopeptidasas/metabolismo , Toxina Shiga/metabolismo , Actinas/metabolismo , Animales , Sistemas de Secreción Bacterianos/fisiología , Línea Celular , Técnica del Anticuerpo Fluorescente , Humanos , Íleon/citología , Íleon/metabolismo , Íleon/ultraestructura , Mucosa Intestinal/ultraestructura , Ratones , Microscopía Electrónica de Transmisión
17.
Metallomics ; 2(9): 596-608, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21072351

RESUMEN

Copper plays an indispensable role in the physiology of the human central nervous system (CNS). As a cofactor of dopamine-ß-hydroxylase, peptidyl-α-monooxygenase, superoxide dismutases, and many other enzymes, copper is a critical contributor to catecholamine biosynthesis, activation of neuropeptides and hormones, protection against reactive oxygen species, respiration and other processes essential for normal CNS function. Copper content in the CNS is tightly regulated, and changes in copper levels in the brain are associated with a wide spectrum of pathologies. However, the mechanistic understanding of copper transport in the CNS is still in its infancy. Little is known about copper distribution among various cell types or cell-specific regulation of copper homeostasis, despite the fact that the molecules mediating copper transport and distribution in the brain (CTR1, Atox1, CCS, ScoI/II, ATP7A and ATP7B) have been identified and their importance in CNS function increasingly understood. In this review, we summarize current knowledge about copper levels and uses in the CNS and describe the molecules involved in maintaining copper homeostasis in the brain.


Asunto(s)
Encéfalo/metabolismo , Cobre/metabolismo , Animales , Cobre/farmacocinética , Humanos , Distribución Tisular
19.
Am J Physiol Gastrointest Liver Physiol ; 296(2): G433-44, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19033537

RESUMEN

ATP7B is a copper-transporting P-type ATPase present predominantly in liver. In basal copper, hepatic ATP7B is in a post-trans-Golgi network (TGN) compartment where it loads cytoplasmic Cu(I) onto newly synthesized ceruloplasmin. When copper levels rise, the protein redistributes via unique vesicles to the apical periphery where it exports intracellular Cu(I) into bile. We want to understand the mechanisms regulating the copper-sensitive trafficking of ATP7B. Earlier, our laboratory reported the presence of apical targeting/TGN retention information within residues 1-63 of human ATP7B; deletion of these residues resulted in a mutant protein that was not efficiently retained in the post-TGN in low copper and constitutively trafficked to the basolateral membrane of polarized, hepatic WIF-B cells with and without copper (13). In this study, we used mutagenesis and adenovirus infection of WIF-B cells followed by confocal immunofluorescence microscopy analysis to identify the precise retention/targeting sequences in the context of full-length ATP7B. We also analyzed the expression of selected mutants in livers of copper-deficient and -loaded mice. Our combined results clearly demonstrate that nine amino acids, F(37)AFDNVGYE(45), comprise an essential apical targeting determinant for ATP7B in elevated copper and participate in the TGN retention of the protein under low-copper conditions. The signal is novel, does not require phosphorylation, and is highly conserved in approximately 24 species of ATP7B. Furthermore, N41S, which is part of the signal we identified, is the first and only Wilson disease-causing missense mutation in residues 1-63 of ATP7B. Expression of N41S-ATP7B in WIF-B cells severely disabled the targeting and retention of the protein. We present a working model of how this physiologically relevant signal might work.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Transporte de Catión/metabolismo , Cobre/metabolismo , Hígado/enzimología , Señales de Clasificación de Proteína , Red trans-Golgi/enzimología , Adenosina Trifosfatasas/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Proteínas de Transporte de Catión/genética , Línea Celular Tumoral , Cobre/deficiencia , Sulfato de Cobre/administración & dosificación , ATPasas Transportadoras de Cobre , Fibroblastos/enzimología , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones , Microscopía Confocal , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación Missense , Transporte de Proteínas , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Transducción Genética
20.
Am J Physiol Gastrointest Liver Physiol ; 294(2): G576-88, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18096610

RESUMEN

Junctional adhesion molecule (JAM) is involved in tight junction (TJ) formation in epithelial cells. Three JAMs (A, B, and C) are expressed in rat hepatocytes, but only rat JAM-A is present in polarized WIF-B cells, a rat-human hepatic line. We used knockdown (KD) and overexpression in WIF-B cells to determine the role of JAM-A in the development of hepatic polarity. Expression of rat JAM-A short hairpin RNA resulted in approximately 50% KD of JAM-A and substantial loss of hepatic polarity, as measured by the absence of apical cysts formed by adjacent cells and sealed by TJ belts. When inhibitory RNA-resistant human JAM-A (huWT) was expressed in KD cells, hepatic polarity was restored. In contrast, expression of JAM-A that either lacked its PDZ-binding motif (huDeltaC-term) or harbored a point mutation (T273A) did not complement, indicating that multiple sites within JAM-A's cytoplasmic tail are required for the development of hepatic polarity. Overexpression of huWT in normal WIF-B cells unexpectedly blocked WIF-B maturation to the hepatic phenotype, as did expression of three huJAM-A constructs with single point mutations in putative phosphorylation sites. In contrast, huDeltaC-term was without effect, and the T273A mutant only partially blocked maturation. Our results show that JAM-A is essential for the development of polarity in cultured hepatic cells via its possible phosphorylation and recruitment of relevant PDZ proteins and that hepatic polarity is achieved within a narrow range of JAM-A expression levels. Importantly, formation/maintenance of TJs and the apical domain in hepatic cells are linked, unlike simple epithelia.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Polaridad Celular/genética , Polaridad Celular/fisiología , Hepatocitos/fisiología , Inmunoglobulinas/fisiología , Secuencia de Aminoácidos , Animales , Moléculas de Adhesión Celular/genética , Línea Celular , Citoplasma/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoglobulinas/genética , Lentivirus/genética , Datos de Secuencia Molecular , Fosforilación , Plásmidos/genética , Proteína Quinasa C/metabolismo , Ratas , Receptores de Superficie Celular , Treonina/metabolismo , Transducción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA