Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood Cells Mol Dis ; 42(3): 233-40, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19380103

RESUMEN

Oligomerization, function, and regulation of unmodified mouse Kcc1 K-Cl cotransporter were studied by chemical crosslinking. Treatment of Xenopus oocytes and 293T cells expressing K-Cl cotransporter Kcc1 with several types of chemical cross-linkers shifted Kcc1 polypeptide to higher molecular weight forms. More extensive studies were performed with the amine-reactive disuccinyl suberate (DSS) and with the sulfhydryl-reactive bis-maleimidohexane (BMH). Kcc1 cross-linking was time-dependent in intact oocytes, and was independent of protein concentration in detergent lysates from oocytes or 293T cells. Kcc1 cross-linking by the cleavable cross-linker DTME was reversible. The N-terminal and C-terminal cytoplasmic tails of Kcc1 were not essential for Kcc1 crosslinking. PFO-PAGE and gel filtration revealed oligomeric states of uncrosslinked KCC1 corresponding in mobility to that of cross-linked protein. DSS and BMH each inhibited KCC1-mediated (86)Rb(+) uptake stimulated by hypotonicity or by N-ethylmaleimide (NEM) without reduction in nominal surface abundance of KCC1. These data add to evidence supporting the oligomeric state of KCC polypeptides.


Asunto(s)
Reactivos de Enlaces Cruzados/farmacología , Simportadores/química , Animales , Línea Celular , Cromatografía en Gel , Electroforesis en Gel de Poliacrilamida , Femenino , Humanos , Transporte Iónico/efectos de los fármacos , Riñón/citología , Riñón/embriología , Ratones , Microscopía Fluorescente , Peso Molecular , Oocitos , Estructura Terciaria de Proteína , ARN Complementario/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/efectos de los fármacos , Radioisótopos de Rubidio/metabolismo , Simportadores/efectos de los fármacos , Xenopus laevis , Cotransportadores de K Cl
2.
J Comp Neurol ; 497(1): 78-87, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16680762

RESUMEN

Recent evidence suggests that substance P (SP) is up-regulated in primary sensory neurons following axotomy and that this change occurs in larger neurons that do not usually produce SP. If this is so, then the up-regulation may allow normally neighboring, uninjured, and nonnociceptive dorsal root ganglion (DRG) neurons to become effective in activating pain pathways. By using immunohistochemistry, we performed a unilateral L5 spinal nerve transection on male Wistar rats and measured SP expression in ipsilateral L4 and L5 DRGs and contralateral L5 DRGs at 1-14 days postoperatively (dpo) and in control and sham-operated rats. In normal and sham-operated DRGs, SP was detectable almost exclusively in small neurons (< or =800 microm2). After surgery, the mean size of SP-positive neurons from the axotomized L5 ganglia was greater at 2, 4, 7, and 14 dpo. Among large neurons (>800 microm2) from the axotomized L5, the percentage of SP-positive neurons increased at 2, 4, 7, and 14 dpo. Among small neurons from the axotomized L5, the percentage of SP-positive neurons was increased at 1 and 3 dpo but was decreased at 7 and 14 dpo. Thus, SP expression is affected by axonal damage, and the time course of the expression is different between large and small DRG neurons. These data support a role for SP-producing, large DRG neurons in persistent sensory changes resulting from nerve injury.


Asunto(s)
Ganglios Espinales/metabolismo , Regulación de la Expresión Génica/fisiología , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/fisiopatología , Sustancia P/metabolismo , Animales , Recuento de Células/métodos , Lateralidad Funcional/fisiología , Ganglios Espinales/patología , Inmunohistoquímica/métodos , Masculino , Neuronas/clasificación , Neuronas/metabolismo , Ratas , Ratas Wistar , Factores de Tiempo
3.
Physiol Genomics ; 8(2): 87-98, 2002 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-11875186

RESUMEN

Polycystin-1 (PKD1) mutations account for approximately 85% of autosomal dominant polycystic kidney disease (ADPKD). We have shown previously that oocyte surface expression of a transmembrane fusion protein encoding part of the cytoplasmic COOH terminus of PKD1 increases activity of a Ca2+-permeable cation channel. We show here that human ADPKD mutations incorporated into this fusion protein attenuated or abolished encoded cation currents. Point mutations and truncations showed that cation current expression requires integrity of a region encompassing the putative coiled coil domain of the PKD1 cytoplasmic tail. Whereas these loss-of-function mutants did not exhibit dominant negative phenotypes, coexpression of a fusion protein expressing the interacting COOH-terminal cytoplasmic tail of PKD2 did suppress cation current. Liganding of the ectodomain of the PKD1 fusion protein moderately activated cation current. The divalent cation permeability and pharmacological profile of the current has been extended. Inducible expression of the PKD1 fusion in EcR-293 cells was also associated with activation of cation channels and increased Ca2+ entry.


Asunto(s)
Canales de Calcio/fisiología , Mutación Missense , Fragmentos de Péptidos/fisiología , Proteínas/fisiología , Animales , Calcio/antagonistas & inhibidores , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Cationes Bivalentes/antagonistas & inhibidores , Cationes Bivalentes/metabolismo , Línea Celular , Citoplasma/genética , Citoplasma/fisiología , Análisis Mutacional de ADN , Humanos , Ligandos , Oocitos/química , Oocitos/citología , Oocitos/metabolismo , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/fisiopatología , Biosíntesis de Proteínas , Estructura Terciaria de Proteína/genética , Estructura Terciaria de Proteína/fisiología , Proteínas/química , Proteínas/genética , Receptores de IgG/biosíntesis , Receptores de IgG/química , Receptores de IgG/genética , Receptores de IgG/fisiología , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología , Canales Catiónicos TRPP , Regulación hacia Arriba/genética , Xenopus laevis/embriología
4.
Am J Physiol Cell Physiol ; 293(2): C738-48, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17652430

RESUMEN

Association of some plasma membrane bicarbonate transporters with carbonic anhydrase enzymes forms a bicarbonate transport metabolon to facilitate metabolic CO(2)-HCO(3)(-) conversions and coupled HCO(3)(-) transport. The transmembrane carbonic anhydrase, CAIX, with its extracellular catalytic site, is highly expressed in parietal and other cells of gastric mucosa, suggesting a role in acid secretion. We examined in transfected HEK293 cells the functional and physical interactions between CAIX and the parietal cell Cl(-)/HCO(3)(-) exchanger AE2 or the putative Cl(-)/HCO(3)(-) exchanger SLC26A7. Coexpression of CAIX increased AE2 transport activity by 28 +/- 7% and also activated transport mediated by AE1 and AE3 (32 +/- 10 and 37 +/- 9%, respectively). In contrast, despite a transport rate comparable to that of AE3, coexpressed CAIX did not alter transport associated with SLC26A7. The CAIX-associated increase of AE2 activity did not result from altered AE2 expression or cell surface processing. CAIX was coimmunoprecipitated with the coexpressed SLC4 polypeptides AE1, AE2, and AE3, but not with SLC26A7. GST pull-down assays with a series of domain-deleted forms of CAIX revealed that the catalytic domain of CAIX mediated interaction with AE2. AE2 and CAIX colocalized in human gastric mucosa, as indicated by coimmunofluorescence. This is the first example of a functional and physical interaction between a bicarbonate transporter and a transmembrane carbonic anhydrase. We conclude that CAIX can bind to some Cl(-)/HCO(3)(-) exchangers to form a bicarbonate transport metabolon.


Asunto(s)
Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Proteínas de Transporte de Anión/metabolismo , Antígenos de Neoplasias/metabolismo , Antiportadores/metabolismo , Bicarbonatos/metabolismo , Anhidrasas Carbónicas/metabolismo , Membrana Celular/metabolismo , Cloruros/metabolismo , Animales , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Proteínas de Transporte de Anión/genética , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Antiportadores/genética , Anhidrasa Carbónica IX , Anhidrasas Carbónicas/química , Anhidrasas Carbónicas/genética , Dominio Catalítico , Línea Celular , Membrana Celular/enzimología , Mucosa Gástrica/enzimología , Mucosa Gástrica/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Mutación , Células Parietales Gástricas/enzimología , Células Parietales Gástricas/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Proteínas SLC4A , Factores de Tiempo , Transfección
5.
J Am Soc Nephrol ; 18(5): 1408-18, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17409310

RESUMEN

Mutations in the human gene that encodes the AE1 Cl(-)/HCO(3)(-) exchanger (SLC4A1) cause autosomal recessive and dominant forms of distal renal tubular acidosis (dRTA). A mouse model that lacks AE1/slc4a1 (slc4a1-/-) exhibited dRTA characterized by spontaneous hyperchloremic metabolic acidosis with low net acid excretion and, inappropriately, alkaline urine without bicarbonaturia. Basolateral Cl(-)/HCO(3)(-) exchange activity in acid-secretory intercalated cells of isolated superfused slc4a1-/- medullary collecting duct was reduced, but alternate bicarbonate transport pathways were upregulated. Homozygous mice had nephrocalcinosis associated with hypercalciuria, hyperphosphaturia, and hypocitraturia. A severe urinary concentration defect in slc4a1-/- mice was accompanied by dysregulated expression and localization of the aquaporin-2 water channel. Mice that were heterozygous for the AE1-deficient allele had no apparent defect. Thus, the slc4a1-/- mouse is the first genetic model of complete dRTA and demonstrates that the AE1/slc4a1 Cl(-)/HCO(3)(-) exchanger is required for maintenance of normal acid-base homeostasis by distal renal regeneration of bicarbonate in the mouse as well as in humans.


Asunto(s)
Acidosis Tubular Renal/genética , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Desequilibrio Ácido-Base/sangre , Desequilibrio Ácido-Base/orina , Acidosis Tubular Renal/sangre , Acidosis Tubular Renal/complicaciones , Acidosis Tubular Renal/orina , Animales , Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Antiportadores/genética , Antiportadores/metabolismo , Femenino , Perfilación de la Expresión Génica , Concentración de Iones de Hidrógeno , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nefrocalcinosis/etiología , Nefrocalcinosis/patología
6.
J Cell Sci ; 118(Pt 22): 5279-89, 2005 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16263761

RESUMEN

Dishevelled family proteins are multidomain intracellular transducers of Wnt signals. Ectopically expressed mammalian Dishevelled 2 (Dvl-2) activates downstream signalling and localises to cytoplasmic puncta. It has been suggested that these Dvl-2-containing structures correspond to intracellular vesicles and may be involved in the Wnt signal transduction process. We report that cytoplasmic puncta are primarily formed in cells expressing Dvl-2 at high levels. Lower levels of expression can activate signalling without forming puncta. The structures do not localise with markers of the early or late endocytic pathway and time-lapse analysis demonstrates that Dvl-2 puncta move in a random fashion over short distances but do not originate from the plasma membrane. Based on our findings, we propose that Dvl-2 puncta are protein aggregates that are not required for signalling.


Asunto(s)
Citoplasma/metabolismo , Fosfoproteínas/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Antígenos CD/metabolismo , Biomarcadores/análisis , Cricetinae , Proteínas Dishevelled , Perros , Endocitosis , Retículo Endoplásmico/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Fosfoproteínas/química , Fosfoproteínas/genética , Glicoproteínas de Membrana Plaquetaria/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Transducción de Señal/efectos de los fármacos , Tetraspanina 30 , Factores de Tiempo , Transfección , Proteínas de Transporte Vesicular
7.
Am J Physiol Renal Physiol ; 289(4): F835-49, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15914778

RESUMEN

Although the zebrafish has been used increasingly for the study of pronephric kidney development, studies of renal ion transporters and channels of the zebrafish remain few. We report the cDNA cloning and characterization of the AE2 anion exchanger ortholog from zebrafish kidney, slc4a2/ae2. The ae2 gene in linkage group 2 encodes a polypeptide of 1,228 aa exhibiting 64% aa identity with mouse AE2a. The exon-intron boundaries of the zebrafish ae2 gene are nearly identical to those of the rodent and human genes. Whole-mount in situ hybridization detects ae2 mRNA in prospective midbrain as early as the five-somite stage, then later in the pronephric primordia and the forming pronephric duct, where it persists through 72 h postfertilization (hpf). Zebrafish Ae2 expressed in Xenopus laevis oocytes mediates Na(+)-independent, electroneutral (36)Cl(-)/Cl(-) exchange moderately sensitive to inhibition by DIDS, is inhibited by acidic intracellular pH and by acidic extracellular pH, but activated by (acidifying) ammonium and by hypertonicity. Zebrafish Ae2 also mediates Cl(-)/HCO(3)(-) exchange in X. laevis oocytes and accumulates in or near the plasma membrane in transfected HEK-293 cells. In 24-48 hpf zebrafish embryos, the predominant but not exclusive localization of Ae2 polypeptide is the apical membrane of pronephric duct epithelial cells. Thus Ae2 resembles its mammalian orthologs in function, mechanism, and acute regulation but differs in its preferentially apical expression in kidney. These results will inform tests of the role of Ae2 in zebrafish kidney development and function.


Asunto(s)
Proteínas de Transporte de Anión/genética , Antiportadores/genética , ADN Complementario/biosíntesis , Secuencia de Aminoácidos , Animales , Bicarbonatos/metabolismo , Western Blotting , Línea Celular , Antiportadores de Cloruro-Bicarbonato , Cloruros/metabolismo , Mapeo Cromosómico , Clonación Molecular , ADN Complementario/genética , Embrión no Mamífero , Exones/genética , Humanos , Hibridación in Situ , Intrones/genética , Datos de Secuencia Molecular , Oocitos/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas SLC4A , Fracciones Subcelulares/metabolismo , Xenopus laevis , Pez Cebra
8.
Am J Physiol Gastrointest Liver Physiol ; 286(2): G312-20, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12958022

RESUMEN

Large volumes of saliva are generated by transepithelial Cl(-) movement during parasympathetic muscarinic receptor stimulation. To gain further insight into a major Cl(-) uptake mechanism involved in this process, we have characterized the anion exchanger (AE) activity in mouse serous parotid and mucous sublingual salivary gland acinar cells. The AE activity in acinar cells was Na(+) independent, electroneutral, and sensitive to the anion exchange inhibitor DIDS, properties consistent with the AE members of the SLC4A gene family. Localization studies using a specific antibody to the ubiquitously expressed AE2 isoform labeled acini in both parotid and sublingual glands. Western blot analysis detected an approximately 170-kDa protein that was more highly expressed in the plasma membranes of sublingual than in parotid glands. Correspondingly, the DIDS-sensitive Cl(-)/HCO(3)(-) exchanger activity was significantly greater in sublingual acinar cells. The carbonic anhydrase antagonist acetazolamide markedly inhibited, whereas muscarinic receptor stimulation enhanced, the Cl(-)/HCO(3)(-) exchanger activity in acinar cells from both glands. Intracellular Ca(2+) chelation prevented muscarinic receptor-induced upregulation of the AE, whereas raising the intracellular Ca(2+) concentration with the Ca(2+)-ATPase inhibitor thapsigargin mimicked the effects of muscarinic receptor stimulation. In summary, carbonic anhydrase activity was essential for regulating Cl(-)/HCO(3)(-) exchange in salivary gland acinar cells. Moreover, muscarinic receptor stimulation enhanced AE activity through a Ca(2+)-dependent mechanism. Such forms of regulation may play important roles in modulating fluid and electrolyte secretion by salivary gland acinar cells.


Asunto(s)
Acetazolamida/farmacología , Proteínas de Transporte de Anión , Antiportadores , Calcio/metabolismo , Inhibidores de Anhidrasa Carbónica/farmacología , Antiportadores de Cloruro-Bicarbonato/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Muscarínicos/fisiología , Glándulas Salivales/metabolismo , Ácido 4,4'-Diisotiocianostilbeno-2,2'-Disulfónico/farmacología , Animales , Anhidrasas Carbónicas/metabolismo , Membranas Intracelulares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Concentración Osmolar , Glándula Parótida/metabolismo , Proteínas SLC4A , Glándulas Salivales/citología , Glándulas Salivales/efectos de los fármacos , Sodio/fisiología , Glándula Sublingual/metabolismo
9.
J Biol Chem ; 278(45): 44949-58, 2003 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-12933803

RESUMEN

Cl-/HCO3- exchange activity mediated by the AE1 anion exchanger is reduced by carbonic anhydrase II (CA2) inhibition or by prevention of CA2 binding to the AE1 C-terminal cytoplasmic tail. This type of AE1 inhibition is thought to represent reduced metabolic channeling of HCO3- to the intracellular HCO3- binding site of AE1. To test the hypothesis that CA2 binding might itself allosterically activate AE1 in Xenopus oocytes, we compared Cl-/Cl- and Cl-/HCO3- exchange activities of AE1 polypeptides with truncation and missense mutations in the C-terminal tail. The distal renal tubular acidosis-associated AE1 901X mutant exhibited both Cl-/Cl- and Cl-/HCO3- exchange activities. In contrast, AE1 896X, 891X, and AE1 missense mutants in the CA2 binding site were inactive as Cl-/HCO3- exchangers despite exhibiting normal Cl-/Cl- exchange activities. Co-expression of CA2 enhanced wild-type AE1-mediated Cl-/HCO3- exchange, but not Cl-/Cl- exchange. CA2 co-expression could not rescue Cl-/HCO3- exchange activity in AE1 mutants selectively impaired in Cl-/HCO3- exchange. However, co-expression of transport-incompetent AE1 mutants with intact CA2 binding sites completely rescued Cl-/HCO3- exchange by an AE1 missense mutant devoid of CA2 binding, with activity further enhanced by CA2 co-expression. The same transport-incompetent AE1 mutants failed to rescue Cl-/HCO3- exchange by the AE1 truncation mutant 896X, despite preservation of the latter's core CA2 binding site. These data increase the minimal extent of a functionally defined CA2 binding site in AE1. The inter-protomeric rescue of HCO3- transport within the AE1 dimer shows functional proximity of the C-terminal cytoplasmic tail of one protomer to the anion translocation pathway in the adjacent protomer within the AE1 heterodimer. The data strongly support the hypothesis that an intact transbilayer anion translocation pathway is completely contained within an AE1 monomer.


Asunto(s)
Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Bicarbonatos/metabolismo , Anhidrasa Carbónica II/metabolismo , Cloruros/metabolismo , Subunidades de Proteína/metabolismo , Secuencia de Aminoácidos , Animales , Proteína 1 de Intercambio de Anión de Eritrocito/química , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Aniones , Sitios de Unión/genética , Transporte Biológico , Anhidrasa Carbónica II/genética , Dimerización , Femenino , Eliminación de Gen , Expresión Génica , Humanos , Membrana Dobles de Lípidos/metabolismo , Ratones , Datos de Secuencia Molecular , Mutagénesis , Mutación Missense , Oocitos/metabolismo , Subunidades de Proteína/química , Subunidades de Proteína/genética , Relación Estructura-Actividad , Transfección , Xenopus
10.
Histochem Cell Biol ; 117(4): 335-44, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11976906

RESUMEN

Potassium-chloride cotransporters (KCCs) encoded by at least four homologous genes are believed to contribute to cell volume regulation and transepithelial ion transport. We have studied KCC polypeptide expression and immunolocalization of KCCs in rat salivary glands and pancreas. Immunoblot analysis of submandibular, parotid, and pancreas plasma membrane fractions with immunospecific antibodies raised against mouse KCC1 revealed protein bands at ca 135 kDa and ca 150 kDa. Immunocytochemical analysis of fixed salivary and pancreas tissue revealed basolateral KCC1 distribution in rat parotid and pancreatic acinar cells, as well as in parotid, submandibular, and pancreatic duct cells. KCC1 or the polypeptide product(s) of one or more additional KCC genes was also expressed in the basolateral membranes of submandibular acinar cells. Both immunoblot and immunofluorescence signals were abolished in the presence of the peptide antigen. These results establish the presence in rat exocrine glands of KCC1 and likely other KCC polypeptides, and suggest a contribution of KCC polypeptides to transepithelial Cl(-) transport.


Asunto(s)
Páncreas/metabolismo , Glándula Parótida/metabolismo , Glándula Submandibular/metabolismo , Simportadores/metabolismo , Animales , Fraccionamiento Celular , Membrana Celular/metabolismo , Células Cultivadas , Técnica del Anticuerpo Fluorescente Indirecta , Immunoblotting , Masculino , Ratones , Páncreas/citología , Glándula Parótida/citología , Péptidos , Ratas , Ratas Wistar , Glándula Submandibular/citología , Simportadores/inmunología , Transfección , Xenopus , Cotransportadores de K Cl
11.
Nephrol Dial Transplant ; 19(2): 371-9, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14736961

RESUMEN

BACKGROUND: Mutations in the human SLC4A1 (AE1/band 3) gene are associated with hereditary spherocytic anaemia and with distal renal tubular acidosis (dRTA). The molecular diagnosis of AE1 mutations has been complicated by the absence of highly polymorphic genetic markers, and the pathogenic mechanisms of some dRTA-associated AE1 mutations remain unclear. Here, we characterized a polymorphic dinucleotide repeat close to the human AE1 gene and performed an immunocytochemical study of kidney tissue from a patient with inherited dRTA with a defined AE1 mutation. METHODS: One CA repeat region was identified in a phage P1-derived artificial chromosome (PAC) clone containing most of the human AE1 gene and the upstream flanking region. We determined its heterozygosity value in multiple populations by PCR analysis. Genotyping of one family with dominant dRTA identified the AE1 R589H mutation, and family member genotypes were compared with the CA repeat length. AE1 and vH(+)-ATPase polypeptides in kidney tissue from an AE1 R589H patient were examined by immunocytochemistry for the first time. RESULTS: This CA repeat, previously reported as D17S1183, is approximately 90 kb upstream of the AE1 gene and displayed considerable length polymorphism, with small racial differences, and a heterozygosity value of 0.56. The allele-specific length of this repeat confirmed co-segregation of the AE1 R589H mutation with the disease phenotype in a family with dominant dRTA. Immunostaining of the kidney cortex from one affected member with superimposed chronic pyelonephritis revealed vH(+)-ATPase-positive intercalated cells in which AE1 was undetectable, and proximal tubular epithelial cells with apparently enhanced apical vH(+)-ATPase staining. CONCLUSIONS: The highly polymorphic dinucleotide repeat adjacent to the human AE1 gene may be useful for future studies of disease association and haplotype analysis. Intercalated cells persist in the end-stage kidney of a patient with familial autosomal dominant dRTA associated with the AE1 R589H mutation. The absence of detectable AE1 polypeptide in those intercalated cells supports the genetic prediction that the AE1 R589H mutation indeed causes dominant dRTA.


Asunto(s)
Acidosis Tubular Renal/genética , Acidosis Tubular Renal/patología , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Predisposición Genética a la Enfermedad , Mutación , Polimorfismo Genético , Acidosis Tubular Renal/epidemiología , Estudios de Casos y Controles , Estudios de Cohortes , Femenino , Regulación de la Expresión Génica , Genes Dominantes , Marcadores Genéticos/genética , Pruebas Genéticas , Humanos , Incidencia , Masculino , Linaje , Valores de Referencia , Sensibilidad y Especificidad
12.
J Biol Chem ; 279(29): 30531-9, 2004 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-15123620

RESUMEN

The AE2 Cl-/HCO3- exchanger is expressed in numerous cell types, including epithelial cells of the kidney, respiratory tract, and alimentary tract. In gastric epithelia, AE2 is particularly abundant in parietal cells, where it may be the predominant mechanism for HCO3- efflux and Cl- influx across the basolateral membrane that is needed for acid secretion. To investigate the hypothesis that AE2 is critical for parietal cell function and to assess its importance in other tissues, homozygous null mutant (AE2(-/-)) mice were prepared by targeted disruption of the AE2 (Slc4a2) gene. AE2(-/-) mice were emaciated, edentulous (toothless), and exhibited severe growth retardation, and most of them died around the time of weaning. AE2(-/-) mice exhibited achlorhydria, and histological studies revealed abnormalities of the gastric epithelium, including moderate dilation of the gastric gland lumens and a reduction in the number of parietal cells. There was little evidence, however, that parietal cell viability was impaired. Ultrastructural analysis of AE2(-/-) gastric mucosa revealed abnormal parietal cell structure, with severely impaired development of secretory canaliculi and few tubulovesicles but normal apical microvilli. These results demonstrate that AE2 is essential for gastric acid secretion and for normal development of secretory canalicular and tubulovesicular membranes in mouse parietal cells.


Asunto(s)
Proteínas de Transporte de Anión , Antiportadores , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Aclorhidria/genética , Alelos , Animales , Northern Blotting , Western Blotting , Supervivencia Celular , Antiportadores de Cloruro-Bicarbonato , Células Epiteliales/metabolismo , Epitelio/metabolismo , Mucosa Gástrica/metabolismo , Vectores Genéticos , Concentración de Iones de Hidrógeno , Membranas Intracelulares/metabolismo , Ratones , Ratones Mutantes , Ratones Transgénicos , Microscopía Confocal , Microscopía Electrónica , Microscopía Fluorescente , Mutación , Células Parietales Gástricas/metabolismo , Fenotipo , ARN Mensajero/metabolismo , Proteínas SLC4A , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA