Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Mol Gastroenterol Hepatol ; 13(2): 599-622, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34610499

RESUMO

BACKGROUND: Autophagosome, the central organelle in autophagy process, can assemble via canonical pathway mediated by LC3-II, the lipidated form of autophagy-related protein LC3/ATG8, or noncanonical pathway mediated by the small GTPase Rab9. Canonical autophagy is essential for exocrine pancreas homeostasis, and its disordering initiates and drives pancreatitis. The involvement of noncanonical autophagy has not been explored. We examine the role of Rab9 in pancreatic autophagy and pancreatitis severity. METHODS: We measured the effect of Rab9 on parameters of autophagy and pancreatitis responses using transgenic mice overexpressing Rab9 (Rab9TG) and adenoviral transduction of acinar cells. Effect of canonical autophagy on Rab9 was assessed in ATG5-deficient acinar cells. RESULTS: Pancreatic levels of Rab9 and its membrane-bound (active) form decreased in rodent pancreatitis models and in human disease. Rab9 overexpression stimulated noncanonical and inhibited canonical/LC3-mediated autophagosome formation in acinar cells through up-regulation of ATG4B, the cysteine protease that delipidates LC3-II. Conversely, ATG5 deficiency caused Rab9 increase in acinar cells. Inhibition of canonical autophagy in Rab9TG pancreas was associated with accumulation of Rab9-positive vacuoles containing markers of mitochondria, protein aggregates, and trans-Golgi. The shift to the noncanonical pathway caused pancreatitis-like damage in acinar cells and aggravated experimental pancreatitis. CONCLUSIONS: The results show that Rab9 regulates pancreatic autophagy and indicate a mutually antagonistic relationship between the canonical/LC3-mediated and noncanonical/Rab9-mediated autophagy pathways in pancreatitis. Noncanonical autophagy fails to substitute for its canonical counterpart in protecting against pancreatitis. Thus, Rab9 decrease in experimental and human pancreatitis is a protective response to sustain canonical autophagy and alleviate disease severity.


Assuntos
Pâncreas , Pancreatite , Células Acinares/metabolismo , Animais , Autofagossomos , Autofagia , Camundongos , Pancreatite/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab de Ligação ao GTP/farmacologia
2.
Sci Adv ; 5(3): eaav8423, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30906870

RESUMO

The urea channel of Helicobacter pylori (HpUreI) is an ideal drug target for preventing gastric cancer but incomplete understanding of its gating mechanism has hampered development of inhibitors for the eradication of H. pylori. Here, we present the cryo-EM structures of HpUreI in closed and open conformations, both at a resolution of 2.7 Å. Our hexameric structures of this small membrane protein (~21 kDa/protomer) resolve its periplasmic loops and carboxyl terminus that close and open the channel, and define a gating mechanism that is pH dependent and requires cooperativity between protomers in the hexamer. Gating is further associated with well-resolved changes in the channel-lining residues that modify the shape and length of the urea pore. Site-specific mutations in the periplasmic domain and urea pore identified key residues important for channel function. Drugs blocking the urea pore based on our structures should lead to a new strategy for H. pylori eradication.


Assuntos
Proteínas de Bactérias/ultraestrutura , Infecções por Helicobacter/microbiologia , Helicobacter pylori/ultraestrutura , Proteínas de Membrana Transportadoras/ultraestrutura , Proteínas de Bactérias/química , Microscopia Crioeletrônica , Infecções por Helicobacter/genética , Helicobacter pylori/química , Helicobacter pylori/genética , Humanos , Concentração de Íons de Hidrogênio , Proteínas de Membrana Transportadoras/química , Conformação Proteica , Neoplasias Gástricas/genética , Neoplasias Gástricas/microbiologia
3.
Sci Rep ; 7(1): 6632, 2017 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-28747707

RESUMO

The gastric proton pump H+,K+-ATPase acidifies the gastric lumen, and thus its inhibitors, including the imidazo[1,2-a]pyridine class of K+-competitive acid blockers (P-CABs), have potential application as acid-suppressing drugs. We determined the electron crystallographic structure of H+,K+-ATPase at 6.5 Å resolution in the E2P state with bound BYK99, a potent P-CAB with a restricted ring structure. The BYK99 bound structure has an almost identical profile to that of a previously determined structure with bound SCH28080, the original P-CAB prototype, but is significantly different from the previously reported P-CAB-free form, illustrating a common conformational change is required for P-CAB binding. The shared conformational changes include a distinct movement of transmembrane helix 2 (M2), from its position in the previously reported P-CAB-free form, to a location proximal to the P-CAB binding site in the present BYK99-bound structure. Site-specific mutagenesis within M2 revealed that D137 and N138, which face the P-CAB binding site in our model, significantly affect the inhibition constant (K i) of P-CABs. We also found that A335 is likely to be near the bridging nitrogen at the restricted ring structure of the BYK99 inhibitor. These provide clues to elucidate the binding site parameters and mechanism of P-CAB inhibition of gastric acid secretion.


Assuntos
Microscopia Crioeletrônica , ATPase Trocadora de Hidrogênio-Potássio/química , Inibidores da Bomba de Prótons/química , Piridinas/química , Animais , Ligação Proteica , Conformação Proteica , Inibidores da Bomba de Prótons/metabolismo , Piridinas/metabolismo , Suínos
5.
Nature ; 493(7431): 255-8, 2013 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-23222544

RESUMO

Half the world's population is chronically infected with Helicobacter pylori, causing gastritis, gastric ulcers and an increased incidence of gastric adenocarcinoma. Its proton-gated inner-membrane urea channel, HpUreI, is essential for survival in the acidic environment of the stomach. The channel is closed at neutral pH and opens at acidic pH to allow the rapid access of urea to cytoplasmic urease. Urease produces NH(3) and CO(2), neutralizing entering protons and thus buffering the periplasm to a pH of roughly 6.1 even in gastric juice at a pH below 2.0. Here we report the structure of HpUreI, revealing six protomers assembled in a hexameric ring surrounding a central bilayer plug of ordered lipids. Each protomer encloses a channel formed by a twisted bundle of six transmembrane helices. The bundle defines a previously unobserved fold comprising a two-helix hairpin motif repeated three times around the central axis of the channel, without the inverted repeat of mammalian-type urea transporters. Both the channel and the protomer interface contain residues conserved in the AmiS/UreI superfamily, suggesting the preservation of channel architecture and oligomeric state in this superfamily. Predominantly aromatic or aliphatic side chains line the entire channel and define two consecutive constriction sites in the middle of the channel. Mutation of Trp 153 in the cytoplasmic constriction site to Ala or Phe decreases the selectivity for urea in comparison with thiourea, suggesting that solute interaction with Trp 153 contributes specificity. The previously unobserved hexameric channel structure described here provides a new model for the permeation of urea and other small amide solutes in prokaryotes and archaea.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Helicobacter pylori/química , Prótons , Ureia/metabolismo , Motivos de Aminoácidos , Cristalografia por Raios X , Concentração de Íons de Hidrogênio , Modelos Moleculares , Multimerização Proteica , Estrutura Secundária de Proteína , Homologia Estrutural de Proteína
6.
J Pharmacol Exp Ther ; 339(2): 412-20, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21828261

RESUMO

Inhibition of the gastric H,K-ATPase by the potassium-competitive acid blocker (P-CAB) 1-[5-(2-fluorophenyl)-1-(pyridin-3-ylsulfonyl)-1H-pyrrol-3-yl]-N-methylmethanamine (TAK-438), is strictly K(+)-competitive with a K(i) of 10 nM at pH 7. In contrast to previous P-CABs, this structure has a point positive charge (pK(a) 9.06) allowing for greater accumulation in parietal cells compared with previous P-CABs [e.g., (8-benzyloxy-2-methyl-imidazo(1,2-a)pyridin-3-yl)acetonitrile (SCH28080), pK(a) 5.6]. The dissociation rate of the compound from the isolated ATPase is slower than other P-CABs, with the t(1/2) being 7.5 h in 20 mM KCl at pH 7. The stoichiometry of binding of TAK-438 to the H,K-ATPase is 2.2 nmol/mg in the presence of Mg-ATP, vanadate, or MgP(i). However, TAK-438 also binds enzyme at 1.3 nmol/mg in the absence of Mg(2+). Modeling of the H,K-ATPase to the homologous Na,K-ATPase predicts a close approach and hydrogen bonding between the positively charged N-methylamino group and the negatively charged Glu795 in the K(+)-binding site in contrast to the planar diffuse positive charge of previous P-CABs. This probably accounts for the slow dissociation and high affinity. The model also predicts hydrogen bonding between the hydroxyl of Tyr799 and the oxygens of the sulfonyl group of TAK-438. A Tyr799Phe mutation resulted in a 3-fold increase of the dissociation rate, showing that this hydrogen bonding also contributes to the slow dissociation rate. Hence, this K(+)-competitive inhibitor of the gastric H,K-ATPase should provide longer-lasting inhibition of gastric acid secretion compared with previous drugs of this class.


Assuntos
Inibidores da Bomba de Prótons , Inibidores da Bomba de Prótons/farmacologia , Pirróis/farmacologia , Estômago/efeitos dos fármacos , Sulfonamidas/farmacologia , Laranja de Acridina/metabolismo , Animais , Corantes Fluorescentes/metabolismo , Refluxo Gastroesofágico/tratamento farmacológico , ATPase Trocadora de Hidrogênio-Potássio/química , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Terapia de Alvo Molecular , Fosforilação/efeitos dos fármacos , Ligação Proteica , Inibidores da Bomba de Prótons/análise , Inibidores da Bomba de Prótons/química , Pirróis/química , Pirróis/metabolismo , Software , Estereoisomerismo , Estômago/enzimologia , Sulfonamidas/química , Sulfonamidas/metabolismo , Suínos
7.
Compr Physiol ; 1(4): 2141-53, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23733700

RESUMO

The gastric H(+),K(+)-ATPase is responsible for gastric acid secretion. This ATPase is composed of two subunits, the catalytic α subunit and the structural ß subunit. The α subunit with molecular mass of about 100 kDa has 10 transmembrane domains and is strongly associated with the ß subunit with a single transmembrane segment and a peptide mass of 35 kDa. Its three-dimensional structure is based on homology modeling and site-directed mutagenesis resulting in a proton extrusion and K(+) reabsorption model. There are three conserved H3O(+)-binding sites in the middle of the membrane domain and H3O(+) secretion depends on a conformational change involving Lys(791) insertion into the second H3O(+) site enclosed by E795, E820, and D824 that allows export of protons at a concentration of 160 mM. K(+) countertransport involves binding to this site after the release of protons with retrograde displacement of Lys(791) and then K(+) transfer to E343 and exit to the cytoplasm. This ATPase is the major therapeutic target in treatment of acid-related diseases and there are several known luminal inhibitors allowing analysis of the luminal vestibule. One class contains the acid-activated covalent, thiophilic proton pump inhibitors, the most effective of current acid-suppressive drugs. Their binding sites and trypsinolysis allowed identification of all ten transmembrane segments of the ATPase. In addition, various K(+)-competitive inhibitors of the ATPase are being developed, with the advantage of complete and rapid inhibition of acid secretion independent of pump activity and allowing further refinement of the structure of the luminal vestibule of the E2 form of this ATPase.


Assuntos
ATPase Trocadora de Hidrogênio-Potássio/química , Sequência de Aminoácidos , Animais , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Humanos , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Potássio/metabolismo , Inibidores da Bomba de Prótons/farmacologia
8.
Biochemistry ; 49(21): 4524-32, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20423050

RESUMO

The antimycotic drug clotrimazole inhibits the function of the gastric H,K-ATPase in a manner similar to that observed for the Na,K-ATPase. Because of the high hydrophobicity of the compound, the interaction between clotrimazole and the ion pump occurs at the membrane domain in the apolar core of the membrane. The enzymatic activity was inhibited with a half-saturating concentration of 5.2 microM. Various partial reactions of the pump cycle were analyzed with the electrochromic styryl dye RH421 that has been widely used to study the transport mechanism of P-type ATPases. We discovered that the interaction of clotrimazole with the H,K-ATPase introduces a single "dead-end" branch added to the Post-Albers scheme in the E(1) state of the pump. In this inhibiting state, the ion binding sites have a significantly enhanced affinity for protons and bind up to two protons even at pH 8.5. Inhibition of the pump can be reversed by a decreased pH or increased K(+) concentrations. The mechanistic proposal that allows an explanation of all experiments presented is similar to that published for the Na,K-ATPase.


Assuntos
Adenosina Trifosfatases/metabolismo , Clotrimazol/metabolismo , Íons/metabolismo , Prótons , Estômago/enzimologia , Animais , Sítios de Ligação , Mucosa Gástrica/metabolismo , Suínos
9.
Biochemistry ; 49(14): 3116-28, 2010 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-20199105

RESUMO

Bulky hydrophilic N-glycans stabilize the proper tertiary structure of glycoproteins. In addition, N-glycans comprise the binding sites for the endoplasmic reticulum (ER)-resident lectins that assist correct folding of newly synthesized glycoproteins. To reveal the role of N-glycans in maturation of the Na,K-ATPase beta(2) subunit in the ER, the effects of preventing or modifying the beta(2) subunit N-glycosylation on trafficking of the subunit and its binding to the ER lectin chaperone, calnexin, were studied in MDCK cells. Preventing N-glycosylation abolishes binding of the beta(2) subunit to calnexin and results in the ER retention of the subunit. Furthermore, the fully N-glycosylated beta(2) subunit is retained in the ER when glycan-calnexin interactions are prevented by castanospermine, showing that N-glycan-mediated calnexin binding is required for correct subunit folding. Calnexin binding persists for several hours after translation is stopped with cycloheximide, suggesting that the beta(2) subunit undergoes repeated post-translational calnexin-assisted folding attempts. Homology modeling of the beta(2) subunit using the crystal structure of the alpha(1)-beta(1) Na,K-ATPase shows the presence of a relatively hydrophobic amino acid cluster proximal to N-glycosylation sites 2 and 7. Combined, but not separate, removal of sites 2 and 7 dramatically impairs calnexin binding and prevents the export of the beta(2) subunit from the ER. Similarly, hydrophilic substitution of two hydrophobic amino acids in this cluster disrupts both beta(2)-calnexin binding and trafficking of the subunit to the Golgi. Therefore, the hydrophobic residues in the proximity of N-glycans 2 and 7 are required for post-translational calnexin binding to these N-glycans in incompletely folded conformers, which, in turn, is necessary for maturation of the Na,K-ATPase beta(2) subunit.


Assuntos
Polissacarídeos/fisiologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Calnexina/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Cães , Retículo Endoplasmático/metabolismo , Glicosilação , Proteínas Luminescentes/genética , Modelos Moleculares , Chaperonas Moleculares/metabolismo , Mutação , Ligação Proteica , Dobramento de Proteína , Processamento de Proteína Pós-Traducional , Subunidades Proteicas/metabolismo , Transporte Proteico , ATPase Trocadora de Sódio-Potássio/genética
10.
Hum Gene Ther ; 20(3): 225-37, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19292604

RESUMO

We used convection-enhanced delivery (CED) to characterize gene delivery mediated by adeno-associated virus type 1 (AAV1) by tracking expression of hrGFP (humanized green fluorescent protein from Renilla reniformis) into the striatum, basal forebrain, and corona radiata of monkey brain. Four cynomolgus monkeys received single infusions into corona radiata, putamen, and caudate. The other group (n = 4) received infusions into basal forebrain. Thirty days after infusion animals were killed and their brains were processed for immunohistochemical evaluation. Volumetric analysis of GFP-positive brain areas was performed. AAV1-hrGFP infusions resulted in approximately 550, 700, and 73 mm(3) coverage after infusion into corona radiata, striatum, and basal forebrain, respectively. Aside from targeted regions, other brain structures also showed GFP signal (internal and external globus pallidus, subthalamic nucleus), supporting the idea that AAV1 is actively trafficked to regions distal from the infusion site. In addition to neuronal transduction, a significant nonneuronal cell population was transduced by AAV1 vector; for example, oligodendrocytes in corona radiata and astrocytes in the striatum. We observed a strong humoral and cell-mediated response against AAV1-hrGFP in transduced monkeys irrespective of the anatomic location of the infusion, as evidenced by induction of circulating anti-AAV1 and anti-hrGFP antibodies, as well as infiltration of CD4(+) lymphocytes and upregulation of MHC-II in regions infused with vector. We conclude that transduction of antigen-presenting cells within the CNS is a likely cause of this response and that caution is warranted when foreign transgenes are used as reporters in gene therapy studies with vectors with broader tropism than AAV2.


Assuntos
Dependovirus/imunologia , Vetores Genéticos/imunologia , Vetores Genéticos/metabolismo , Prosencéfalo/imunologia , Prosencéfalo/metabolismo , Transdução Genética/métodos , Animais , Formação de Anticorpos , Astrócitos/metabolismo , Dependovirus/genética , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Proteínas de Fluorescência Verde , Humanos , Infusões Parenterais , Macaca fascicularis , Doenças do Sistema Nervoso/imunologia , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/terapia , Neurônios/metabolismo , Oligodendroglia/metabolismo , Prosencéfalo/citologia , Proteínas Recombinantes/biossíntese
11.
Pflugers Arch ; 457(3): 609-22, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18536934

RESUMO

The gastric H,K-ATPase, a member of the P(2)-type ATPase family, is the integral membrane protein responsible for gastric acid secretion. It is an alpha,beta-heterodimeric enzyme that exchanges cytoplasmic hydronium with extracellular potassium. The catalytic alpha subunit has ten transmembrane segments with a cluster of intramembranal carboxylic amino acids located in the middle of the transmembrane segments TM4, TM5,TM6, and TM8. Comparison to the known structure of the SERCA pump, mutagenesis, and molecular modeling has identified these as constituents of the ion binding domain. The beta subunit has one transmembrane segment with N terminus in cytoplasmic region. The extracellular domain of the beta subunit contains six or seven N-linked glycosylation sites. N-glycosylation is important for the enzyme assembly, maturation, and sorting. The enzyme pumps acid by a series of conformational changes from an E(1) (ion site in) to an E(2) (ion site out) configuration following binding of MgATP and phosphorylation. Several experimental observations support the hypothesis that expulsion of the proton at 160 mM (pH 0.8) results from movement of lysine 791 into the ion binding site in the E(2)P configuration. Potassium access from the lumen depends on activation of a K and Cl conductance via a KCNQ1/KCNE2 complex and Clic6. K movement through the luminal channel in E(2)P is proposed to displace the lysine along with dephosphorylation to return the enzyme to the E(1) configuration. This enzyme is inhibited by the unique proton pump inhibitor class of drug, allowing therapy of acid-related diseases.


Assuntos
Hormônios Gastrointestinais , Estômago/enzimologia , Sítios de Ligação , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Hormônios Gastrointestinais/antagonistas & inibidores , Hormônios Gastrointestinais/química , Hormônios Gastrointestinais/metabolismo , Humanos , Modelos Moleculares , Estrutura Molecular , Potássio/metabolismo , Conformação Proteica , Estômago/citologia
12.
Biophys J ; 95(6): 2739-49, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18567633

RESUMO

Gastric H,K-ATPase is an electroneutral transmembrane pump that moves protons from the cytoplasm of the parietal cell into the gastric lumen in exchange for potassium ions. The mechanism of transport against the established electrochemical gradients includes intermediate conformations in which the transferred ions are trapped (occluded) within the membrane domain of the pump. The pump cycle involves switching between the E1 and E2P states. Molecular dynamics simulations on homology models of the E2P and E1 states were performed to investigate the mechanism of K(+) movement in this enzyme. We performed separate E2P simulations with one K(+) in the luminal channel, one K(+) ion in the occlusion site, two K(+) ions in the occlusion site, and targeted molecular dynamics from E2P to E1 with two K(+) ions in the occlusion site. The models were inserted into a lipid bilayer system and were stable over the time course of the simulations, and K(+) ions in the channel moved to a consistent location near the center of the membrane domain, thus defining the occlusion site. The backbone carbonyl oxygen from residues 337 through 342 on the nonhelical turn of M4, as well as side-chain oxygen from E343, E795, and E820, participated in the ion occlusion. A single water molecule was stably bound between the two K(+) ions in the occlusion site, providing an additional ligand and partial shielding the positive charges from one another. Targeted molecular dynamics was used to transform the protein from the E2P to the E1 state (two K(+) ions to the cytoplasm). This simulation identified the separation of the water column in the entry channel as the likely gating mechanism on the luminal side. A hydrated exit channel also formed on the cytoplasmic side of the occlusion site during this simulation. Hence, water molecules became available to hydrate the ions. The movement of the M1M2 transmembrane segments, and the displacement of residues Q159, E160, Q110, and T152 during the conformational change, as well as the motions of E343 and L346, acted as the cytoplasmic-side gate.


Assuntos
ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Ativação do Canal Iônico , Modelos Moleculares , Estômago/enzimologia , Citoplasma/metabolismo , ATPase Trocadora de Hidrogênio-Potássio/química , Movimento , Potássio/metabolismo , Conformação Proteica , Água/metabolismo
13.
Mol Ther ; 15(12): 2114-23, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17667945

RESUMO

Non-human primates (NHPs) are considered to be among the most relevant animal models for pre-clinical testing of human therapies, on the basis of their close evolutionary relatedness to humans in terms of organ cell biology and physiology. In this study, we sought to investigate whether NHP models accurately reflect the effectiveness of recombinant adeno-associated virus (rAAV)-mediated gene delivery to the airway in humans. In order to do this, we utilized an identical model system of differentiated airway epithelia from Indian Rhesus monkeys and from humans, cultured at an air-liquid interface (ALI). In addition to assessing the biology of rAAV-mediated transduction for three serotypes, we characterized the bioelectric properties as a reference for biological similarities and differences between the cell cultures from the two species. Our results demonstrate that airway epithelia from NHPs and humans have very similar Na(+) and Cl(-) transport properties. In contrast, rAAV transduction of airway epithelia of NHPs demonstrated significant differences to those in humans with regard to the efficiency of apical and/or basal transduction with three rAAV serotypes (AAV1, AAV2, AAV5). These findings suggest that the IndianRhesusmonkey may not be the best model for preclinical testing of rAAV-mediated gene therapy to the airway in humans.


Assuntos
Brônquios/metabolismo , Dependovirus/genética , Transdução Genética , Animais , Sequência de Bases , Brônquios/citologia , Brônquios/ultraestrutura , Cloretos/metabolismo , Primers do DNA , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Humanos , Macaca mulatta , Microscopia Eletrônica de Varredura , Reação em Cadeia da Polimerase , Recombinação Genética
14.
J Clin Gastroenterol ; 41 Suppl 2: S226-42, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17575528

RESUMO

The recent progress in therapy if acid disease has relied heavily on the performance of drugs targeted against the H,K ATPase of the stomach and the H2 receptor antagonists. It has become apparent in the last decade that the proton pump is the target that has the likelihood of being the most sustainable area of therapeutic application in the regulation of acid suppression. The process of activation of acid secretion requires a change in location of the ATPase from cytoplasmic tubules into the microvilli of the secretory canaliculus of the parietal cell. Stimulation of the resting parietal cell, with involvement of F-actin and ezrin does not use significant numbers of SNARE proteins, because their message is depleted in the pure parietal cell transcriptome. The cell morphology and gene expression suggest a tubule fusion-eversion event. As the active H,K ATPase requires efflux of KCl for activity we have, using the transcriptome derived from 99% pure parietal cells and immunocytochemistry, provided evidence that the KCl pathway is mediated by a KCQ1/KCNE2 complex for supplying K and CLIC6 for supplying the accompanying Cl. The pump has been modeled on the basis of the structures of different conformations of the sr Ca ATPase related to the catalytic cycle. These models use the effects of site directed mutations and identification of the binding domain of the K competitive acid pump antagonists or the defined site of binding for the covalent class of proton pump inhibitors. The pump undergoes conformational changes associated with phosphorylation to allow the ion binding site to change exposure from cytoplasmic to luminal exposure. We have been able to postulate that the very low gastric pH is achieved by lysine 791 motion extruding the hydronium ion bound to carboxylates in the middle of the membrane domain. These models also allow description of the K entry to form the K liganded form of the enzyme and the reformation of the ion site inward conformation thus relating the catalytic cycle of the pump to conformational models. The mechanism of action of the proton pump inhibitor class of drug is discussed along with the cysteines covalently bound with these inhibitors. The review concludes with a discussion of the mechanism of action and binding regions of a possible new class of drug for acid control, the K competitive acid pump antagonists.


Assuntos
Antiulcerosos/farmacologia , Inibidores Enzimáticos/farmacologia , Ácido Gástrico/metabolismo , Mucosa Gástrica/efeitos dos fármacos , Inibidores da Bomba de Prótons , Animais , Antiulcerosos/química , Antiulcerosos/metabolismo , Ligação Competitiva , Domínio Catalítico , Cloretos/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Mucosa Gástrica/enzimologia , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Humanos , Modelos Moleculares , Estrutura Molecular , Células Parietais Gástricas/química , Células Parietais Gástricas/efeitos dos fármacos , Células Parietais Gástricas/enzimologia , Potássio/metabolismo , Ligação Proteica , Conformação Proteica , Transporte Proteico , Relação Estrutura-Atividade
15.
Biochemistry ; 46(18): 5398-417, 2007 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-17425287

RESUMO

New models of the gastric H,K ATPase in the E1K and E2P states are presented as the first structures of a K+ counter-transport P2-type ATPase exhibiting ion entry and exit paths. Homology modeling was first used to generate a starting conformation from the srCa ATPase E2P form (PDB code 1wpg) that contains bound MgADP. Energy minimization of the model showed a conserved adenosine site but nonconserved polyphosphate contacts compared to the srCa ATPase. Molecular dynamics was then employed to expand the luminal entry sufficiently to allow access of the rigid K+ competitive naphthyridine inhibitor, Byk99, to its binding site within the membrane domain. The new E2P model had increased separation between transmembrane segments M3 through M8, and addition of water in this space showed not only an inhibitor entry path to the luminal vestibule but also a channel leading to the ion binding site. Addition of K+ to the hydrated channel with molecular dynamics modeling of ion movement identified a pathway for K+ from the lumen to the ion binding site to give E2K. A K+ exit path to the cytoplasm operating during the normal catalytic cycle is also proposed on the basis of an E1K homology model derived from the E12Ca2+ form of the srCa ATPase (PDB code 1su4). Autodock analyses of the new E2P model now correctly discriminate between high- and low-affinity K+ competitive inhibitors. Finally, the expanded luminal vestibule of the E2P model explains high-affinity ouabain binding in a mutant of the H,K ATPase [Qiu et al. (2005) J. Biol. Chem. 280, 32349-32355].


Assuntos
Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Mucosa Gástrica/enzimologia , ATPase Trocadora de Hidrogênio-Potássio/química , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Transdução de Sinais/fisiologia , Substituição de Aminoácidos/genética , Animais , Sítios de Ligação/genética , Ligação Competitiva/genética , Simulação por Computador , Mucosa Gástrica/metabolismo , ATPase Trocadora de Hidrogênio-Potássio/genética , Transporte de Íons/genética , Modelos Moleculares , Conformação Proteica , Inibidores da Bomba de Prótons , Coelhos , Ratos , Homologia de Sequência de Aminoácidos , Transdução de Sinais/genética , Homologia Estrutural de Proteína , Suínos
16.
J Gene Med ; 9(1): 10-21, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17154340

RESUMO

BACKGROUND: Cystic fibrosis is an autosomal recessive disease affecting approximately 1 in 2500 live births. Introducing the cDNA that codes for normal cystic fibrosis transmembrane conductance regulator (CFTR) to the small airways of the lung could result in restoring the CFTR function. A number of vectors for lung gene therapy have been tried and adeno-associated virus (AAV) vectors offer promise. The vector is delivered to the lung using a breath-actuated jet nebulizer. The purpose of this project was to determine the aerosolized AAV (tgAAVCF) particle size distribution (PSD) in order to calculate target doses for lung delivery. METHODS: A tgAAVCF solution was nebulized using the Pari LC Plus (n = 3), and the PSD was determined by coupling laser diffraction and inertial impaction (NGI) techniques. The NGI allowed for quantification of the tgAAVCF at each stage of impaction, ensuring that rAAV-CFTR vector is present and not empty particles. Applying the results to mathematical algorithms allowed for the calculation of expected pulmonary deposition. RESULTS: The mass median diameter (MMD) for the tgAAVCF was 2.78 +/- 0.43 microm. If the system works ideally and the patient only receives aerosol on inspiration, the patient would receive 47 +/- 0% of the initial dose placed in the nebulizer, with 72 +/- 0.73% of this being deposited beyond the vocal cords. CONCLUSIONS: This technology for categorizing the pulmonary delivery system for lung gene therapy vectors can be adapted for advanced aerosol delivery systems or other vectors.


Assuntos
Aerossóis/farmacocinética , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Pulmão , Administração por Inalação , Albuterol/administração & dosagem , Fibrose Cística/terapia , Dependovirus/genética , Humanos , Lasers , Nebulizadores e Vaporizadores , Tamanho da Partícula
17.
Biochemistry ; 44(14): 5267-84, 2005 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-15807521

RESUMO

The gastric H,K-ATPase catalyzes electroneutral exchange of H(+) for K(+) as a function of enzyme phosphorylation and dephosphorylation during transition between E(1)/E(1)-P (ion site in) and E(2)-P/E(2) (ion site out) conformations. Here we present homology modeling of the H,K-ATPase in the E(2)-P conformation as a means of predicting the interaction of the enzyme with two known classes of specific inhibitors. All known proton pump inhibitors, PPIs, form a disulfide bond with cysteine 813 that is accessible from the luminal surface. This allows allocation of the binding site to a luminal vestibule adjacent to Cys813 enclosed by part of TM4 and the loop between TM5 and TM6. K(+) competitive imidazo-1,2alpha-pyridines also bind to the luminal surface of the E(2)-P conformation, and their binding excludes PPI reaction. This overlap of the binding sites of the two classes of inhibitors combined with the results of site-directed mutagenesis and cysteine cross-linking allowed preliminary assignment of a docking mode for these reversible compounds in a position close to Glu795 that accounts for the detailed structure/activity relationships known for these compounds. The new E(2)-P model is able to assign a possible mechanism for acid secretion by this P(2)-type ATPase. Several ion binding side chains identified in the sr Ca-ATPase by crystallography are conserved in the Na,K- and H,K-ATPases. Poised in the middle of these, the H,K-ATPase substitutes lysine in place of a serine implicated in K(+) binding in the Na,K-ATPase. Molecular models for hydronium binding to E(1) versus E(2)-P predict outward displacement of the hydronium bound between Asp824, Glu820, and Glu795 by the R-NH(3)(+) of Lys791 during the conformational transition from E(1)P and E(2)P. The site for luminal K(+) binding at low pH is proposed to be between carbonyl oxygens in the nonhelical part of the fourth membrane span and carboxyl oxygens of Glu795 and Glu820. This site of K(+) binding is predicted to destabilize hydrogen bonds between these carboxylates and the -NH(3)(+) group of Lys791, allowing the Lys791 side chain to return to its E(1) position.


Assuntos
Inibidores Enzimáticos/metabolismo , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Estômago/enzimologia , Sequência de Aminoácidos , Sítios de Ligação , Western Blotting , Catálise , ATPase Trocadora de Hidrogênio-Potássio/química , ATPase Trocadora de Hidrogênio-Potássio/genética , Íons , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Conformação Proteica , Inibidores da Bomba de Prótons , Homologia de Sequência de Aminoácidos
18.
Mol Ther ; 10(6): 990-1002, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15564131

RESUMO

Pharmacologic- and gene-based therapies have historically been developed as two independent therapeutic platforms for cystic fibrosis (CF) lung disease. Inhibition of the dysregulated epithelial Na channel (ENaC) is one pharmacologic approach to enhance airway clearance in CF. We investigated pharmacologic approaches to enhance CFTR gene delivery with recombinant adeno-associated virus (rAAV) and identified compounds that significantly improved viral transduction while simultaneously inhibiting ENaC activity through an unrelated mechanism. Treatment of human CF airway epithelia with proteasome modulating agents (LLnL and doxorubicin) at the time of rAAV2 or rAAV2/5 infection dramatically enhanced CFTR gene delivery and correction of CFTR-mediated short-circuit currents. Surprisingly, these agents also facilitated long-term (15-day) functional inhibition of ENaC currents independent of CFTR vector administration. Inhibition of ENaC activity was predominantly attributed to a doxorubicin-dependent decrease in gamma-ENaC subunit mRNA expression and an increase in gamma-ENaC promoter methylation. This is the first report to describe the identification of compounds with dual therapeutic action that are able to enhance the efficacy of CFTR gene therapy to the airway while simultaneously ameliorating primary aspects of CF disease pathophysiology. The identification of such compounds mark a new area for drug development, not only for CF, but also for other gene therapy disease targets.


Assuntos
Fibrose Cística/genética , Fibrose Cística/terapia , Doxorrubicina/farmacologia , Terapia Genética/métodos , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma , Amilorida/farmacologia , Polaridade Celular , Células Cultivadas , Ilhas de CpG/genética , Fibrose Cística/tratamento farmacológico , Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Metilação de DNA , Dependovirus/genética , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Canais Epiteliais de Sódio , Genoma Viral , Humanos , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sódio/metabolismo , Canais de Sódio/genética , Canais de Sódio/metabolismo , Fatores de Tempo
19.
J Virol ; 78(6): 2863-74, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14990705

RESUMO

Tripeptidyl aldehyde proteasome inhibitors have been shown to effectively increase viral capsid ubiquitination and transduction of recombinant adeno-associated virus type 2 (rAAV-2) and rAAV-5 serotypes. In the present study we have characterized a second class of proteasome-modulating agents (anthracycline derivatives) for their ability to induce rAAV transduction. The anthracycline derivatives doxorubicin and aclarubicin were chosen for analysis because they have been shown to interact with the proteasome through a mechanism distinct from that of tripeptidyl aldehydes. Our studies demonstrated that doxorubicin and aclarubicin also significantly augmented rAAV transduction in airway cell lines, polarized human airway epithelia, and mouse lungs. Both tripeptidyl aldehyde and anthracycline proteasome-modulating agents similarly augmented nuclear accumulation of rAAV in A549 and IB3 airway cell lines. However, these two cell types demonstrated cell specificity in the ability of N-acetyl-L-leucyl-L-leucyl-L-norleucine (LLnL) or doxorubicin to augment rAAV transduction. Interestingly, the combined administration of LLnL and doxorubicin resulted in substantially increased transduction (>2,000-fold) following apical infection of human polarized epithelia with either rAAV-2 or rAAV-5. In summary, the cell type specificity of LLnL and doxorubicin to induce rAAV transduction, together with the ability of these compounds to synergistically enhance rAAV transduction in polarized airway epithelial induction, suggests that these two classes of compounds likely modulate different proteasome functions that affect rAAV transduction. Findings from this study provide new insights into how modulation of proteasome function can be effectively used to augment rAAV transduction in airway epithelia for gene therapy of cystic fibrosis.


Assuntos
Dependovirus/efeitos dos fármacos , Células Epiteliais/virologia , Complexos Multienzimáticos/antagonistas & inibidores , Transdução Genética , Aclarubicina/farmacologia , Animais , Linhagem Celular , Polaridade Celular , Cisteína Endopeptidases , Inibidores de Cisteína Proteinase/farmacologia , Fibrose Cística , Dependovirus/genética , Dependovirus/patogenicidade , Doxorrubicina/farmacologia , Terapia Genética , Humanos , Leupeptinas/farmacologia , Pulmão/citologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Parvoviridae/virologia , Complexo de Endopeptidases do Proteassoma , Recombinação Genética
20.
Ann N Y Acad Sci ; 986: 106-10, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12763782

RESUMO

We have used homology molecular modeling based on the srCaATPase E(2) conformation, pdb1kju, to predict side chains involved in docking the K(+) competitive inhibitor, SCH28080, to the H,K-ATPase. A model for SCH28080 binding between residues L809 and A335 in the same space utilized by omeprazole is proposed. We also describe modeling MgATP binding to the E(1) structure of the srATPase, pdb1eul, as a paradigm for the Na,K- and H,K-ATPases. The resulting model, E(1).MgATP, visualizes a conformation not yet available by crystallization and successfully predicts a range of published results, including backbone cleavages near V440 (N domain) and V712 (P domain) mediated by FeATP in the Na,K-ATPase. A separate model for MgATP docked to E(2) (pdb1kju) shows that access of the gamma phosphate to D351 is blocked by the A domain. The E(2). MgATP model explains FeATP-mediated cleavages of the Na,K-ATPase near V440 and E214 (A domain) and homologous results in the H,K-ATPase.


Assuntos
Trifosfato de Adenosina/metabolismo , ATPases Transportadoras de Cálcio/metabolismo , Inibidores Enzimáticos/farmacologia , ATPase Trocadora de Hidrogênio-Potássio/química , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Imidazóis/farmacologia , Trifosfato de Adenosina/química , Animais , Sítios de Ligação , ATPases Transportadoras de Cálcio/antagonistas & inibidores , ATPases Transportadoras de Cálcio/química , Mucosa Gástrica/enzimologia , Humanos , Modelos Moleculares , Conformação Molecular , Conformação Proteica/efeitos dos fármacos , Estrutura Secundária de Proteína , Inibidores da Bomba de Prótons , Coelhos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...