Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 482(7383): 98-102, 2012 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-22266938

RESUMO

Hypertension affects one billion people and is a principal reversible risk factor for cardiovascular disease. Pseudohypoaldosteronism type II (PHAII), a rare Mendelian syndrome featuring hypertension, hyperkalaemia and metabolic acidosis, has revealed previously unrecognized physiology orchestrating the balance between renal salt reabsorption and K(+) and H(+) excretion. Here we used exome sequencing to identify mutations in kelch-like 3 (KLHL3) or cullin 3 (CUL3) in PHAII patients from 41 unrelated families. KLHL3 mutations are either recessive or dominant, whereas CUL3 mutations are dominant and predominantly de novo. CUL3 and BTB-domain-containing kelch proteins such as KLHL3 are components of cullin-RING E3 ligase complexes that ubiquitinate substrates bound to kelch propeller domains. Dominant KLHL3 mutations are clustered in short segments within the kelch propeller and BTB domains implicated in substrate and cullin binding, respectively. Diverse CUL3 mutations all result in skipping of exon 9, producing an in-frame deletion. Because dominant KLHL3 and CUL3 mutations both phenocopy recessive loss-of-function KLHL3 mutations, they may abrogate ubiquitination of KLHL3 substrates. Disease features are reversed by thiazide diuretics, which inhibit the Na-Cl cotransporter in the distal nephron of the kidney; KLHL3 and CUL3 are expressed in this location, suggesting a mechanistic link between KLHL3 and CUL3 mutations, increased Na-Cl reabsorption, and disease pathogenesis. These findings demonstrate the utility of exome sequencing in disease gene identification despite the combined complexities of locus heterogeneity, mixed models of transmission and frequent de novo mutation, and establish a fundamental role for KLHL3 and CUL3 in blood pressure, K(+) and pH homeostasis.


Assuntos
Proteínas de Transporte/genética , Proteínas Culina/genética , Hipertensão/genética , Mutação/genética , Pseudo-Hipoaldosteronismo/genética , Desequilíbrio Hidroeletrolítico/genética , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Animais , Sequência de Bases , Pressão Sanguínea/genética , Proteínas de Transporte/química , Estudos de Coortes , Proteínas Culina/química , Eletrólitos , Éxons/genética , Feminino , Perfilação da Expressão Gênica , Genes Dominantes/genética , Genes Recessivos/genética , Genótipo , Homeostase/genética , Humanos , Concentração de Íons de Hidrogênio , Hipertensão/complicações , Hipertensão/fisiopatologia , Masculino , Camundongos , Proteínas dos Microfilamentos , Modelos Moleculares , Dados de Sequência Molecular , Fenótipo , Potássio/metabolismo , Pseudo-Hipoaldosteronismo/complicações , Pseudo-Hipoaldosteronismo/fisiopatologia , Cloreto de Sódio/metabolismo , Desequilíbrio Hidroeletrolítico/complicações , Desequilíbrio Hidroeletrolítico/fisiopatologia
2.
Nat Genet ; 33(2): 125-7, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12539048

RESUMO

Autosomal dominant distal renal tubular acidosis (ddRTA) is caused by mutations in SLC4A1, which encodes the polytopic chloride-bicarbonate exchanger AE1 that is normally expressed at the basolateral surface of alpha-intercalated cells in the distal nephron. Here we report that, in contrast with many disorders in which mutant membrane proteins are retained intracellularly and degraded, ddRTA can result from aberrant targeting of AE1 to the apical surface.


Assuntos
Acidose Tubular Renal/genética , Proteína 1 de Troca de Ânion do Eritrócito/genética , Células Epiteliais/metabolismo , Genes Dominantes , Mutação , Transporte Proteico/fisiologia , Acidose Tubular Renal/metabolismo , Proteína 1 de Troca de Ânion do Eritrócito/metabolismo , Antígenos CD8/imunologia , Caderinas/metabolismo , Células Cultivadas , Células Epiteliais/citologia , Hemaglutininas/imunologia , Humanos , Rim/metabolismo , Fragmentos de Peptídeos/metabolismo
3.
Nat Genet ; 36(4): 400-4, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15052268

RESUMO

ARC syndrome (OMIM 208085) is an autosomal recessive multisystem disorder characterized by neurogenic arthrogryposis multiplex congenita, renal tubular dysfunction and neonatal cholestasis with bile duct hypoplasia and low gamma glutamyl transpeptidase (gGT) activity. Platelet dysfunction is common. Affected infants do not thrive and usually die in the first year of life. To elucidate the molecular basis of ARC, we mapped the disease to a 7-cM interval on 15q26.1 and then identified germline mutations in the gene VPS33B in 14 kindreds with ARC. VPS33B encodes a homolog of the class C yeast vacuolar protein sorting gene, Vps33, that contains a Sec1-like domain important in the regulation of vesicle-to-target SNARE complex formation and subsequent membrane fusion.


Assuntos
Artrogripose/genética , Colestase/genética , Nefropatias/genética , Fusão de Membrana/fisiologia , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Mutação , Proteínas/genética , Proteínas de Transporte Vesicular , Western Blotting , Linhagem Celular , Cromossomos Humanos Par 15 , Eletroforese em Gel de Poliacrilamida , Feminino , Humanos , Masculino , Fusão de Membrana/genética , Proteínas de Membrana/química , Plasmídeos , Proteínas/química , Proteínas SNARE , Síndrome
4.
Am J Physiol Renal Physiol ; 300(1): F157-66, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20980406

RESUMO

The mammalian kidney isoform of the essential chloride-bicarbonate exchanger AE1 differs from its erythrocyte counterpart, being shorter at its N terminus. It has previously been reported that the glycolytic enzyme GAPDH interacts only with erythrocyte AE1, by binding to the portion not found in the kidney isoform. (Chu H, Low PS. Biochem J 400:143-151, 2006). We have identified GAPDH as a candidate binding partner for the C terminus of both AE1 and AE2. We show that full-length AE1 and GAPDH coimmunoprecipitated from both human and rat kidney as well as from Madin-Darby canine kidney (MDCK) cells stably expressing kidney AE1, while in human liver, AE2 coprecipitated with GAPDH. ELISA and glutathione S-transferase (GST) pull-down assays using GST-tagged C-terminal AE1 fusion protein confirmed that the interaction is direct; fluorescence titration revealed saturable binding kinetics with Kd 2.3±0.2 µM. Further GST precipitation assays demonstrated that the D902EY residues in the D902EYDE motif located within the C terminus of AE1 are important for GAPDH binding. In vitro GAPDH activity was unaffected by C-terminal AE1 binding, unlike in erythrocytes. Also, differently from red cell N-terminal binding, GAPDH-AE1 C-terminal binding was not disrupted by phosphorylation of AE1 in kidney AE1-expressing MDCK cells. Importantly, small interfering RNA knockdown of GAPDH in these cells resulted in significant intracellular retention of AE1, with a concomitant reduction in AE1 at the cell membrane. These results indicate differences between kidney and erythrocyte AE1/GAPDH behavior and show that in the kidney, GAPDH is required for kidney AE1 to achieve stable basolateral residency.


Assuntos
Proteína 1 de Troca de Ânion do Eritrócito/metabolismo , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Rim/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Transporte de Ânions/metabolismo , Antiporters/metabolismo , Glutationa Transferase/metabolismo , Humanos , Dados de Sequência Molecular , Ratos , Proteínas SLC4A
5.
Hum Mol Genet ; 18(16): 2963-74, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19465746

RESUMO

Familial juvenile hyperuricaemic nephropathy (FJHN), an autosomal dominant disorder, is caused by mutations in the UMOD gene, which encodes Uromodulin, a glycosylphosphatidylinositol-anchored protein that is expressed in the thick ascending limb of the loop of Henle and excreted in the urine. Uromodulin contains three epidermal growth factor (EGF)-like domains, a cysteine-rich region which includes a domain of eight cysteines and a zona pellucida (ZP) domain. Over 90% of UMOD mutations are missense, and 62% alter a cysteine residue, implicating a role for protein misfolding in the disease. We investigated 20 northern European FJHN probands for UMOD mutations. Wild-type and mutant Uromodulins were functionally studied by expression in HeLa cells and by the use of western blot analysis and confocal microscopy. Six different UMOD missense mutations (Cys32Trp, Arg185Gly, Asp196Asn, Cys217Trp, Cys223Arg and Gly488Arg) were identified. Patients with UMOD mutations were phenotypically similar to those without UMOD mutations. The mutant Uromodulins had significantly delayed maturation, retention in the endoplasmic reticulum (ER) and reduced expression at the plasma membrane. However, Gly488Arg, which is the only mutation we identified in the ZP domain, was found to be associated with milder in vitro abnormalities and to be the only mutant Uromodulin detected in conditioned medium from transfected cells, indicating that the severity of the mutant phenotypes may depend on their location within the protein. Thus, FJHN-causing Uromodulin mutants are retained in the ER, with impaired intracellular maturation and trafficking, thereby indicating mechanisms whereby Uromodulin mutants may cause the phenotype of FJHN.


Assuntos
Retículo Endoplasmático/metabolismo , Hiperuricemia/genética , Mucoproteínas/genética , Mutação de Sentido Incorreto , Adolescente , Adulto , Idoso , Retículo Endoplasmático/química , Retículo Endoplasmático/genética , Feminino , Células HeLa , Humanos , Hiperuricemia/metabolismo , Masculino , Pessoa de Meia-Idade , Mucoproteínas/química , Mucoproteínas/metabolismo , Linhagem , Dobramento de Proteína , Transporte Proteico , Uromodulina , População Branca/genética , Adulto Jovem
6.
Nephron Physiol ; 118(1): p28-34, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21071985

RESUMO

Disorders of water balance lead either to dehydration or overhydration. Because there is an intimate relationship between water and sodium concentration (water generally following salt), one can distinguish hypotonic, isotonic and hypertonic dehydration and the same for overhydration. The vast majority of water balance disorders are acquired. In this article, the focus is on the inherited disorders both of water (nephrogenic diabetes insipidus) and acid-base balance. Both acidosis and alkalosis can arise from primary tubular ion transport abnormalities. The alkaloses are usually secondary to salt handling problems, whereas the renal tubular acidoses are often a consequence of primary abnormalities of acid-base transporters.


Assuntos
Desequilíbrio Ácido-Base/genética , Síndrome de Fanconi/genética , Predisposição Genética para Doença/genética , Homeostase/genética , Nefropatias/genética , Túbulos Renais Proximais/fisiopatologia , Animais , Genótipo , Humanos , Modelos Genéticos
7.
J Am Soc Nephrol ; 20(2): 251-4, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19193780

RESUMO

The form of renal tubular acidosis associated with hyperkalemia is usually attributable to real or apparent hypoaldosteronism. It is therefore a common feature in diabetes and a number of other conditions associated with underproduction of renin or aldosterone. In addition, the close relationship between potassium levels and ammonia production dictates that hyperkalemia per se can lead to acidosis. Here I describe the modern relationship between molecular function of the distal portion of the nephron, pathways of ammoniagenesis, and hyperkalemia.


Assuntos
Acidose Tubular Renal/fisiopatologia , Hiperpotassemia/fisiopatologia , Túbulos Renais/patologia , Acidose/patologia , Acidose Tubular Renal/metabolismo , Aldosterona/metabolismo , Amônia/metabolismo , Animais , Canais Epiteliais de Sódio/metabolismo , Humanos , Hiperpotassemia/metabolismo , Modelos Biológicos , Néfrons/patologia , Potássio/metabolismo
8.
Curr Opin Nephrol Hypertens ; 18(5): 433-8, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19561496

RESUMO

PURPOSE OF REVIEW: Vacuolar-type H+ATPases are multisubunit macromolecules that play an essential role in renal acid-base homeostasis. Other cellular processes also rely on the proton pumping ability of H+ATPases to acidify organellar or lumenal spaces. Several diseases, including distal renal tubular acidosis, osteoporosis and wrinkly skin syndrome, are due to mutations in genes encoding alternate subunits that make up the H+ATPase. This review highlights recent key articles in this research area. RECENT FINDINGS: Further insights into the structure, expression and regulation of H+ATPases have been elucidated, within the kidney and elsewhere. This knowledge may enhance the potential for future drug targeting. SUMMARY: Novel findings concerning tissue-specific subunits of the H+ATPase that are important in the kidney and more general lessons of H+ATPase function and regulation are slowly emerging, though the paucity of cellular tools available has to date limited progress.


Assuntos
Rim/enzimologia , ATPases Translocadoras de Prótons/fisiologia , Animais , Membrana Celular/enzimologia , Endocitose/fisiologia , Humanos , Rim/crescimento & desenvolvimento , Nefropatias/enzimologia , ATPases Translocadoras de Prótons/biossíntese , ATPases Translocadoras de Prótons/química
9.
Gene ; 393(1-2): 94-100, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17350184

RESUMO

Several of the 13 subunits comprising mammalian H(+)-ATPases have multiple alternative forms, encoded by separate genes and with differing tissue expression patterns. These may play an important role in the intracellular localization and activity of H(+)-ATPases. Here we report the cloning of a previously uncharacterized human gene, ATP6V0E2, encoding a novel H(+)-ATPase e-subunit designated e2. We demonstrate that in contrast to the ubiquitously expressed gene encoding the e1 subunit (previously called e), this novel gene is expressed in a more restricted tissue distribution, particularly kidney and brain. We show by complementation studies in a yeast strain deficient for the ortholog of this subunit, that either form of the e-subunit is essential for proper proton pump function. The identification of this novel form of the e-subunit lends further support to the hypothesis that subunit differences may play a key role in the structure, site and function of H(+)-ATPases within the cell.


Assuntos
Subunidades Proteicas/genética , Bombas de Próton/genética , ATPases Vacuolares Próton-Translocadoras/genética , Acidose Tubular Renal/enzimologia , Acidose Tubular Renal/genética , Processamento Alternativo/genética , Sequência de Aminoácidos , Clonagem Molecular , DNA Complementar/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Teste de Complementação Genética , Humanos , Dados de Sequência Molecular , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , ATPases Vacuolares Próton-Translocadoras/química , ATPases Vacuolares Próton-Translocadoras/metabolismo , Leveduras/crescimento & desenvolvimento
10.
J Clin Invest ; 113(8): 1075-81, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15085183

RESUMO

The kidney plays a central role in our ability to maintain appropriate sodium balance, which is critical to determination of blood pressure. In this review we outline current knowledge of renal salt handling at the molecular level, and, given that Westernized societies consume more salt than is required for normal physiology, we examine evidence that the lowering of salt intake can combat hypertension.


Assuntos
Hipertensão/etiologia , Sódio na Dieta/administração & dosagem , Sódio/metabolismo , Alcalose/genética , Animais , Síndrome de Bartter/genética , Humanos , Hipertensão/genética , Hipertensão/metabolismo , Rim/metabolismo , Pseudo-Hipoaldosteronismo/genética
11.
Gene ; 297(1-2): 169-77, 2002 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-12384298

RESUMO

Several of the 13 subunits comprising mammalian H(+)-ATPases have multiple isoforms, encoded by separate genes and with differing tissue expression patterns, which may play an important role in the intracellular localization and activity of H(+)-ATPases. Here we report the cloning of three previously uncharacterized human genes, ATP6V1C2, ATP6V1G3 and ATP6V0D2, encoding novel H(+)-ATPase subunit isoforms C2, G3 and d2, respectively. We demonstrate that these novel genes are expressed in kidney and few other tissues, and confirm previous reports that the C1, G1 and d1 isoforms are ubiquitously expressed, while G2 is brain-specific. Previously we have shown that mutations in two kidney-specific genes, ATP6V1B1 and ATP6V0A4, encoding the H(+)-ATPase B1 and a4 subunit isoforms, cause recessive distal renal tubular acidosis (dRTA). As the genes reported here are expressed mainly in kidney, we assessed their candidacy as causative genes for recessive dRTA in eight kindreds unlinked to either known disease locus. Although no potential disease-causing mutations were seen in this cohort, this does not rule out a role for these genes in other families. The identification of these three novel tissue-specific isoforms supports the hypothesis that subunit differences may play a key role in the structure, site and function of H(+)-ATPases within the cell.


Assuntos
Acidose Tubular Renal/genética , Rim/enzimologia , ATPases Vacuolares Próton-Translocadoras/genética , Processamento Alternativo , Sequência de Aminoácidos , Clonagem Molecular , DNA Complementar/química , DNA Complementar/genética , Saúde da Família , Regulação Enzimológica da Expressão Gênica , Genes Recessivos , Humanos , Interações Hidrofóbicas e Hidrofílicas , Isoenzimas/química , Isoenzimas/genética , Rim/metabolismo , Dados de Sequência Molecular , Mutação , Subunidades Proteicas , Alinhamento de Sequência , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , ATPases Vacuolares Próton-Translocadoras/química
12.
J Histochem Cytochem ; 52(10): 1377-84, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15385584

RESUMO

The endolymph in the endolymphatic sac (ES) is acidic (pH 6.6-7). Maintaining this acidic lumen is believed to be important for the normal function of the ES. The acid-base regulation mechanisms of the ES are unknown. Here we investigated the expression patterns of acid-base regulators, including vacuolar (v)H+-ATPase (proton pump), carbonic anhydrase (CA) II, and pendrin in the murine ES epithelium by immunohistochemistry (IHC) and compared their expression patterns by double immunostaining. We found that pendrin and vH+-ATPase were co-localized in the apical membrane of a specific type of ES epithelial cell. Pendrin- and vH+-ATPase-positive cells also expressed cytoplasmic CA II. Co-expression of pendrin, vH+-ATPase, and CA II in the same subgroup of ES cells suggests that this specific type of ES cell is responsible for the acid-base balance processes in the ES and pendrin, vH+-ATPase, and CA II are involved in these processes.


Assuntos
Anidrase Carbônica II/biossíntese , Proteínas de Transporte/biossíntese , Saco Endolinfático/enzimologia , Células Epiteliais/enzimologia , Proteínas de Membrana Transportadoras , ATPases Vacuolares Próton-Translocadoras/biossíntese , Animais , Saco Endolinfático/citologia , Imunofluorescência , Imuno-Histoquímica , Camundongos , Subunidades Proteicas/biossíntese , Transportadores de Sulfato
13.
J Nephrol ; 15 Suppl 6: S57-68, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12515375

RESUMO

In this review I will briefly discuss two major categories of inherited renal tubular transport disorders: those primarily affecting sodium and potassium (whether retention or wasting), and the primary renal acidopathies. The kidney does not of course carry out any of its myriad homeostatic functions in a vacuum, and interactions with other systems form an important part of the overall picture. This is perhaps most marked in the context of salt homeostasis, where the endocrine system, particularly the mineralocorticoid axis and aldosterone, are closely dovetailed. While these disorders are in general rare, the insights gained in understanding their molecular bases have added significantly to our understanding of normal human physiology. The hope is that these findings may in the future prove relevant to the understanding and treatment of more common disorders.


Assuntos
Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/fisiopatologia , Erros Inatos do Transporte Tubular Renal/genética , Erros Inatos do Transporte Tubular Renal/fisiopatologia , Doenças Genéticas Inatas/terapia , Humanos , Túbulos Renais/fisiopatologia , Erros Inatos do Transporte Tubular Renal/terapia , ATPase Trocadora de Sódio-Potássio/genética , ATPase Trocadora de Sódio-Potássio/fisiologia
14.
J Biomol Tech ; 22(4): 136-45, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22131889

RESUMO

Advances in mass spectrometry (MS) have encouraged interest in its deployment in urine biomarker studies, but success has been limited. Urine exosomes have been proposed as an ideal source of biomarkers for renal disease. However, the abundant urinary protein, uromodulin, cofractionates with exosomes during isolation and represents a practical contaminant that limits MS sensitivity. Uromodulin depletion has been attempted but is labor- and time-intensive and may remove important protein biomarkers. We describe the application of an exclusion list (ExL) of uromodulin-related peptide ions, coupled with high-sensitivity mass spectrometric analysis, to increase the depth of coverage of the urinary exosomal proteome. Urine exosomal protein samples from healthy volunteers were subjected to tandem MS and abundant uromodulin peptides identified. Samples were run for a second time, while excluding these uromodulin peptides from fragmentation to allow identification of peptides from lower-abundance proteins. Uromodulin exclusion was performed in addition to dynamic exclusion. Results from these two procedures revealed 222 distinct proteins from conventional analysis, compared with 254 proteins after uromodulin exclusion, of which 188 were common to both methods. By unmasking a previously unidentified protein set, adding the ExL increased overall protein identifications by 29.7% to a total of 288 proteins. A fixed ExL, used in combination with conventional methods, effectively increases the depth of urinary exosomal proteins identified by MS, reducing the need for uromodulin depletion.


Assuntos
Biomarcadores/urina , Proteoma/análise , Proteoma/química , Espectrometria de Massas em Tandem/métodos , Uromodulina/análise , Sequência de Aminoácidos , Cromatografia Líquida , Exossomos/química , Humanos , Dados de Sequência Molecular , Peptídeos/química , Proteômica/métodos , Uromodulina/química
15.
Int Med Case Rep J ; 4: 7-11, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-23754897

RESUMO

Autosomal recessive distal renal tubular acidosis is usually a severe disease of childhood, often presenting as failure to thrive in infancy. It is often, but not always, accompanied by sensorineural hearing loss, the clinical severity and age of onset of which may be different from the other clinical features. Mutations in either ATP6V1B1 or ATP6V0A4 are the chief causes of primary distal renal tubular acidosis with or without hearing loss, although the loss is often milder in the latter. We describe a kindred with compound heterozygous alterations in ATP6V0A4, where hearing loss was formally diagnosed late in both siblings such that they missed early opportunities for hearing support. This kindred highlights the importance of routine audiologic assessments of all children with distal renal tubular acidosis, irrespective either of age at diagnosis or of which gene is mutated. In addition, when diagnostic genetic testing is undertaken, both genes should be screened irrespective of current hearing status. A strategy for this is outlined.

16.
PLoS One ; 5(3): e9531, 2010 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-20224822

RESUMO

The vacuolar-type H(+)-ATPase (V-ATPase) is a multisubunit proton pump that is involved in both intra- and extracellular acidification processes throughout the body. Multiple homologs and splice variants of V-ATPase subunits are thought to explain its varied spatial and temporal expression pattern in different cell types. Recently subunit nomenclature was standardized with a total of 22 subunit variants identified. However this standardization did not accommodate the existence of splice variants and is therefore incomplete. Thus, we propose here an extension of subunit nomenclature along with a literature and sequence database scan for additional V-ATPase subunits. An additional 17 variants were pulled from a literature search while 4 uncharacterized potential subunit variants were found in sequence databases. These findings have been integrated with the current V-ATPase knowledge base to create a new V-ATPase subunit catalogue. It is envisioned this catalogue will form a new platform on which future studies into tissue- and organelle-specific V-ATPase expression, localization and function can be based.


Assuntos
Processamento Alternativo , ATPases Vacuolares Próton-Translocadoras/química , ATPases Vacuolares Próton-Translocadoras/genética , Algoritmos , Animais , Bases de Dados Genéticas , Genoma Humano , Humanos , RNA Mensageiro/metabolismo , Terminologia como Assunto , Fatores de Tempo
18.
Am J Physiol Renal Physiol ; 296(6): F1279-90, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19357182

RESUMO

Rhesus glycoprotein homologs RhAG, RhBG, and RhCG comprise a recently identified branch of the Mep/Amt ammonia transporter family. Animal studies have shown that RhBG and RhCG are present in the kidney distal tubules. Studies in mouse and rat tissue suggest a basolateral localization for RhBG in cells of the distal tubules including the alpha-intercalated cells (alpha-IC), but no localization of RhBG has been reported in human tissue. To date RhCG localization has been described as exclusively apical plasma membrane in mouse and rat kidney, or apical and basolateral in humans, and some mouse and rat tissue studies. We raised novel antibodies to RhBG and RhCG to examine their localization in the human kidney. Madin-Darby canine kidney (MDCKI) cell lines stably expressing human green fluorescent protein-tagged RhBG or RhCG and human tissue lysates were used to demonstrate the specificity of these antibodies for detecting RhBG and RhCG. Using immunoperoxidase staining and antigen liberation techniques, both apical and basolateral RhCG localization was observed in the majority of the cells of the distal convoluted tubule and IC of the connecting tubule and collecting duct. Confocal microscopic imaging of normal human kidney cryosections showed that RhCG staining was predominantly localized to the apical membrane in these cells with some basolateral and intracellular staining evident. A proportion of RhCG staining labeled kAE1-positive cells, confirming that RhCG is localized to the alpha-IC cells. Surprisingly, no RhBG protein was detectable in the human kidney by Western blot analysis of tissue lysates, or by immunohistochemistry or confocal microscopy of tissue sections. The same antibodies, however, could detect RhBG in rat tissue. We conclude that under normal conditions, RhCG is the major putative ammonia transporter expressed in the human kidney and RhBG is not expressed at detectable levels.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Regulação da Expressão Gênica/fisiologia , Glicoproteínas/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Animais , Anticorpos/imunologia , Especificidade de Anticorpos , Proteínas de Transporte de Cátions/genética , Linhagem Celular , Cães , Glicoproteínas/genética , Humanos , Rim/citologia , Rim/metabolismo , Glicoproteínas de Membrana/genética , Proteínas de Membrana Transportadoras/genética , ATPase Trocadora de Sódio-Potássio/metabolismo
19.
J Bioenerg Biomembr ; 40(4): 371-80, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18752060

RESUMO

The multi-subunit vacuolar-type H(+)-ATPase consists of a V(1) domain (A-H subunits) catalyzing ATP hydrolysis and a V(0) domain (a, c, c', c", d, e) responsible for H(+) translocation. The mammalian V(0) d subunit is one of the least-well characterized, and its function and position within the pump are still unclear. It has two different forms encoded by separate genes, d1 being ubiquitous while d2 is predominantly expressed at the cell surface in kidney and osteoclast. To determine whether it forms part of the pump's central stalk as suggested by bacterial A-ATPase studies, or is peripheral as hypothesized from a yeast model, we investigated both human d subunit isoforms. In silico structural modelling demonstrated that human d1 and d2 are structural orthologues of bacterial subunit C, despite poor sequence identity. Expression studies of d1 and d2 showed that each can pull down the central stalk's D and F subunits from human kidney membrane, and in vitro studies using D and F further showed that the interactions between these proteins and the d subunit is direct. These data indicate that the d subunit in man is centrally located within the pump and is thus important in its rotary mechanism.


Assuntos
Rim/enzimologia , Modelos Químicos , ATPases Translocadoras de Prótons/química , ATPases Translocadoras de Prótons/metabolismo , Vacúolos/enzimologia , Sítios de Ligação , Simulação por Computador , Ativação Enzimática , Estabilidade Enzimática , Humanos , Modelos Moleculares , Ligação Proteica , Subunidades Proteicas , ATPases Translocadoras de Prótons/ultraestrutura , Relação Estrutura-Atividade
20.
Am J Physiol Renal Physiol ; 295(4): F950-8, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18632794

RESUMO

The vacuolar-type ATPase (H+ATPase) is a ubiquitously expressed multisubunit pump whose regulation is poorly understood. Its membrane-integral a-subunit is involved in proton translocation and in humans has four forms, a1-a4. This study investigated two naturally occurring point mutations in a4's COOH terminus that cause recessive distal renal tubular acidosis (dRTA), R807Q and G820R. Both lie within a domain that binds the glycolytic enzyme phosphofructokinase-1 (PFK-1). We recreated these disease mutations in yeast to investigate effects on protein expression, H+ATPase assembly, targeting and activity, and performed in vitro PFK-1 binding and activity studies of mammalian proteins. Mammalian studies revealed complete loss of binding between the COOH terminus of a4 containing the G-to-R mutant and PFK-1, without affecting PFK-1's catalytic activity. In yeast expression studies, protein levels, H+ATPase assembly, and targeting of this mutant were all preserved. However, severe (78%) loss of proton transport but less decrease in ATPase activity (36%) were observed in mutant vacuoles, suggesting a requirement for the a-subunit/PFK-1 binding to couple these two functions. This role for PFK in H+ATPase function was supported by similar functional losses and uncoupling ratio between the two proton pump domains observed in vacuoles from a PFK-null strain, which was also unable to grow at alkaline pH. In contrast, the R-to-Q mutation dramatically reduced a-subunit production, abolishing H+ATPase function completely. Thus in the context of dRTA, stability and function of the metabolon composed of H+ATPase and glycolytic components can be compromised by either loss of required PFK-1 binding (G820R) or loss of pump protein (R807Q).


Assuntos
Acidose Tubular Renal/fisiopatologia , Fosfofrutoquinase-1/metabolismo , ATPases Translocadoras de Prótons/genética , ATPases Translocadoras de Prótons/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Acidose Tubular Renal/metabolismo , Sequência de Aminoácidos , Dicroísmo Circular , Glicólise/fisiologia , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fosfofrutoquinase-1/genética , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/genética , Ressonância de Plasmônio de Superfície , ATPases Vacuolares Próton-Translocadoras
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA