首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 0 毫秒
1.
Ontogeny of the Neutral Amino Acid Transporter SNAT1 in the Developing Rat   总被引:2,自引:0,他引:2  
Summary System A is a highly regulated, Na+-dependent transporter that accepts neutral amino acids containing short, polar side chains. System A plays an important role during rat development as decreased pup weights are observed in dams infused during gestation with a non-metabolizable System A substrate. Given the potential importance of SNAT1 during development in the rat brain, we examined whether SNAT1 would be present at an earlier gestation during organogenesis in multiple organs by immunohistochemistry and immunoblotting. SNAT1 protein was observed in the developing lungs, intestines, kidneys, heart, pancreas, and skeletal muscle of rats at prenatal days 14, 17, 19, 21, and postnatal day 2 rats. SNAT1 protein expression decreased in the liver and intestine shortly after birth and as the rat matured. SNAT1 expression was constant throughout development in the lungs and kidney and increased in the heart from prenatal day 19 to postnatal day 2. Highest levels of expression in older animals were seen in organs undergoing rapid cell division.  相似文献   

2.
In previous work, we found that gain-of-function mutations that hyperactivate GEM-1 (an SLC16A transporter protein) can bypass the requirement for GON-2 (a TRPM channel protein) during the initiation of gonadogenesis in C. elegans. Consequently, we proposed that GEM-1 might function as part of a Mg2+ uptake pathway that functions in parallel to GON-2. In this study, we report that CATP-6, a C. elegans ortholog of the P5B ATPase, ATP13A2 (PARK9), is necessary for gem-1 gain-of-function mutations to suppress the effects of gon-2 inactivation. One possible explanation for this observation is that GEM-1 serves to activate CATP-6, which then functions as a Mg2+ transporter. However, we found that overexpression of GEM-1 can alleviate the requirement for CATP-6 activity, suggesting that CATP-6 probably acts as a non-essential upstream positive regulator of GEM-1. Our results are consistent with the notion that P5B ATPases govern intracellular levels of Mg2+ and/or Mn2+ by regulating the trafficking of transporters and other proteins associated with the plasma membrane.  相似文献   

3.
Phenolic acids such asp-coumaric acid and microbial metabolites of poorly absorbed polyphenols are absorbed by the monocarboxylic acid transporter (MCT)-mediated transport system which is identical to the fluorescein/H+ cotransport system. We focus here on the physiological impact of MCT-mediated absorption and distribution. We examined whether MCT1, the best-characterized isoform found in almost all tissues, is involved in this MCT-mediated transport system. The induction of MCT1 expression in Caco-2 cells by a treatment with sodium butyrate (NaBut) did not increase the fluorescein permeability. Moreover, the transfection of Caco-2 cells with an expression vector encoding MCT1 caused no increase in either the permeability or uptake of fluorescein. Furthermore, in the MCT1-expressing oocytes, no increase ofp-coumaric acid uptake was apparent, whereas the uptake of salicylic acid, a substrate of MCT1, nearly doubled. Our data therefore establish that MCT1 was not involved in the MCT-mediated transport of phenolic acids.  相似文献   

4.
The high yield expression of the human LAT1 transporter has been obtained for the first time using E. coli. The hLAT1 cDNA was amplified from HEK293 cells and cloned in pH6EX3 vector. The construct pH6EX3-6His-hLAT1 was used to express the 6His-hLAT1 protein in the Rosetta(DE3)pLysS strain of E. coli. The highest level of expression was detected 8 h after induction by IPTG at 28 °C. The expressed protein was collected in the insoluble fraction of cell lysate. On SDS-PAGE the apparent molecular mass of the polypeptide was 40 kDa. After solubilization with sarkosyl and denaturation with urea the protein carrying a 6His N-terminal tag was purified by Ni2+-chelating affinity chromatography and identified by anti-His antibody. The yield of the over-expressed protein after purification was 3.5 mg/L (cell culture). The human CD98 cDNA amplified from Imagene plasmid was cloned in pGEX-4T1. The construct pGEX-4T1-hCD98 was used to express the GST-hCD98 protein in the Rosetta(DE3)pLysS strain of E. coli. The highest level of expression was detected in this case 4 h after induction by IPTG at 28 °C. The expressed protein was accumulated in the soluble fraction of cell lysate. The molecular mass was determined on the basis of marker proteins on SDS-PAGE; it was about 110 kDa. GST was cleaved from the protein construct by incubation with thrombin for 12 h and the hCD98 was separated by Sephadex G-200 chromatography (size exclusion). hCD98 showed a 62 kDa apparent molecular mass, as determined on the basis of molecular mass markers using SDS-PAGE. The yield of CD98 was 2 mg/L of cell culture.  相似文献   

5.
The L-type amino acid transporter-1 (LAT1, SLC7A5) is upregulated in a wide range of human cancers, positively correlated with the biological aggressiveness of tumors, and a promising target for both imaging and therapy. Radiolabeled amino acids such as O-(2-[18F]fluoroethyl)-L-tyrosine (FET) that are transport substrates for system L amino acid transporters including LAT1 have met limited success for oncologic imaging outside of the brain, and thus new strategies are needed for imaging LAT1 in systemic cancers. Here, we describe the development and biological evaluation of a novel zirconium-89 labeled antibody, [89Zr]DFO-Ab2, targeting the extracellular domain of LAT1 in a preclinical model of colorectal cancer. This tracer demonstrated specificity for LAT1 in vitro and in vivo with excellent tumor imaging properties in mice with xenograft tumors. PET imaging studies showed high tumor uptake, with optimal tumor-to-non target contrast achieved at 7 days post administration. Biodistribution studies demonstrated tumor uptake of 10.5 ± 1.8 percent injected dose per gram (%ID/g) at 7 days with a tumor to muscle ratio of 13 to 1. In contrast, the peak tumor uptake of the radiolabeled amino acid [18F]FET was 4.4 ± 0.5 %ID/g at 30 min after injection with a tumor to muscle ratio of 1.4 to 1. Blocking studies with unlabeled anti-LAT1 antibody demonstrated a 55% reduction of [89Zr]DFO-Ab2 accumulation in the tumor at 7 days. These results are the first report of direct PET imaging of LAT1 and demonstrate the potential of immunoPET agents for imaging specific amino acid transporters.  相似文献   

6.
The SLC38 family of solute transporters mediates the coupled transport of amino acids and Na+ into or out of cells. The structural basis for this coupled transport process is not known. Here, a profile-based sequence analysis approach was used, predicting a distant relationship with the SLC5/6 transporter families. Homology models using the LeuTAa and Mhp1 transporters of known structure as templates were established, predicting the location of a conserved Na+ binding site in the center of membrane helices 1 and 8. This homology model was tested experimentally in the SLC38 member SNAT2 by analyzing the effect of a mutation to Thr-384, which is predicted to be part of this Na+ binding site. The results show that the T384A mutation not only inhibits the anion leak current, which requires Na+ binding to SNAT2, but also dramatically lowers the Na+ affinity of the transporter. This result is consistent with a previous analysis of the N82A mutant transporter, which has a similar effect on anion leak current and Na+ binding and which is also expected to form part of the Na+ binding site. In contrast, random mutations to other sites in the transporter had little or no effect on Na+ affinity. Our results are consistent with a cation binding site formed by transmembrane helices 1 and 8 that is conserved among the SLC38 transporters as well as among many other bacterial and plant transporter families of unknown structure, which are homologous to SLC38.The sodium-coupled neutral amino acid transporter, SNAT2,2 belongs to the SLC38 gene family of solute carrier proteins (1). Together with SNAT1 and -4 (2), it is believed to mediate Na+-dependent amino acid transport activity that was classically assigned to System A transporters (38). In addition to SNAT1 and -2, the SLC38 family has four other known members, two of which predominantly mediate glutamine transport (SNAT3 and -5, System N (911)). SNAT2 is widely expressed in mammalian tissue (1, 7), but it may play a particularly critical role in the brain (12), where it may help shuttle glutamine from astrocytes to neurons via the glutamate-glutamine cycle (1). This process is essential for recycling the neurotransmitter glutamate (13). However, the exact contribution of SNAT2 to the glutamate-glutamine cycle is still controversially discussed (14).Despite this physiological importance, surprisingly little is known about the functional properties and the structural basis of amino acid transport by the SLC38 proteins. Although hydropathy analysis predicts 11 transmembrane helices (TMs), with an intracellular N terminus and an extracellular C terminus (1), it is not clear whether the transporters belong to a large superfamily of transporters, of which members have been characterized structurally through x-ray crystallography. At present, sequence homology has only been established with transporters of the mammalian SLC32 and SLC36 families as well as with the more distantly related plant auxin carriers and the bacterial amino acid-polyamine-organocation (APC) family (15, 16). High resolution crystal structures are not available for any of the transporters from these families, although low resolution projection structures were recently reported for the APC family members AdiC (17) and SteT (18). However, these structures do not allow the assignment of transmembrane helices. Thus, it remains unknown whether the SLC38 fold is similar to established transport protein folds, although homology to the major facilitator superfamily seems unlikely.We have recently identified a conserved amino acid residue in SNAT2, Asn-82, which is involved in controlling the Na+ affinity of the transporter (19). Interestingly, Asn-82 is localized in the predicted TM1 of SNAT2. This first transmembrane helix was recently found to contribute ligands to a Na+ binding site in several bacterial transporters, which are related to the SLC5 (sodium glucose symporter) and SLC6 (sodium- and chloride-dependent neurotransmitter transporter) family members (2022), which also comprises bacterial members (23, 24). Although sequence similarity with SLC5 and -6 is not detectable, SLC38 may be a member of a possibly very large superfamily with the same general fold, which also contains many amino acid transport proteins.Here, we used a homology modeling approach based on profile-based sequence alignment (25, 26). A search against sequences deposited in the Protein Data Bank (PDB (27)) revealed that the transporters with the highest likelihood to share an analogous fold are a leucine transporter from Aquifex aeolicus, LeuTAa, and a homologous hydantoin transporter from Microbacterium liquefaciens, Mhp1. We established a homology model based on these structures, which predicts Asn-82 to be part of a Na+ binding site. Furthermore, another conserved hydrophilic amino acid residue in TM8, Thr-384, was predicted to be near this cation binding site. When Thr-384 was mutated to alanine, a dramatic loss of the affinity of SNAT2 for Na+ was observed, whereas mutations to other sites that were spatially removed from the predicted Na+ binding site had little or no effect on Na+ affinity. We hypothesize that the SLC38 family is a member of a large superfamily of cation/organic substrate transporters which includes the mammalian SLC5 and -6 proteins and which has a conserved cation binding site formed by TMs 1 and 8.  相似文献   

7.
The proton-coupled amino acid transporter 1 (PAT1, SLC36A1) mediates the uptake of small neutral amino acids at the apical membrane of intestinal epithelial cells after protein digestion. The transporter is currently under intense investigation, because it is a possible vehicle for oral drug delivery. Structural features of the protein such as the number of transmembrane domains, the substrate binding site, or essential amino acids are still unknown. In the present study we use mutagenesis experiments and biochemical approaches to determine the role of the three putative extracellular cysteine residues on transport function and their possible involvement in the formation of a disulfide bridge. As treatment with the reducing reagent dithiothreitol impaired transport function of hPAT1 wild type protein, substitution of putative extracellular cysteine residues Cys-180, Cys-329, and Cys-473 by alanine or serine was performed. Replacement of the two highly conserved cysteine residues Cys-180 and Cys-329 abolished the transport function of hPAT1 in Xenopus laevis oocytes. Studies of wild type and mutant transporters expressed in human retinal pigment epithelial (HRPE) cells suggested that the binding of the substrate was inhibited in these mutants. Substitution of the third putative extracellular nonconserved cysteine residue Cys-473 did not affect transport function. All mutants were expressed at the plasma membrane. Biotinylation of free sulfhydryl groups using maleimide-PEG11-biotin and SDS-PAGE analysis under reducing and nonreducing conditions provided direct evidence for the existence of an essential disulfide bond between Cys-180 and Cys-329. This disulfide bridge is very likely involved in forming or stabilizing the substrate binding site.The solute carrier (SLC)2 superfamily represents the second largest group of membrane proteins after the G-protein-coupled receptor (GPCR) superfamily in the human genome. Comprising 384 members, the 46 SLC families include transporters for inorganic ions, amino acids, neurotransmitters, sugars, purines, fatty acids, and other substances (1). Ten SLC families contain 47 known transporters for amino acids and 48 related orphan transporters. Phylogenetic analysis revealed four main clusters (α, β, γ, and δ). Together with members of the SLC32 and SLC38 families, the proton-coupled amino acid transporter 1 (PAT1) was placed into group β. PAT1 is a member of the SLC36 family (SLC36A1). It was originally identified as the lysosomal amino acid transporter (LYAAT1) in rat brain (2). Subsequently, mouse and human homologs were cloned from mouse intestine (3) and from Caco-2 cells (4), respectively. PAT1 is identical to the H+/amino acid cotransporter that has been functionally described in Caco-2 cells (5). It is localized mainly to the apical membrane of intestine epithelial cells and is also found in lysosomes in brain neurons (4) facilitating the transport of amino acids from luminal protein digestion or lysosomal proteolysis, respectively. The transport of substrates via PAT1 is driven by an inwardly directed H+ gradient. Recently we could identify the conserved His-55 as being responsible for binding and translocation of the proton (6).Prototypic substrates for PAT1 are small neutral amino acids (e.g. l-proline, glycine, β-alanine) and amino acid derivatives (e.g. γ-aminobutyric acid (GABA), α-(methylamino)-isobutyric acid) (35, 710). Recently, PAT1 gained much interest because it transports pharmaceutically relevant compounds such as d-cycloserine, l-azetidine-2-carboxylic acid, 3-amino-1-propanesulfonic acid, 3,4-dehydro-l-proline, vigabatrin, and other GABA analogs (8, 10, 11) rendering it an interesting target for the pharmaceutical industry. PAT1 seems to be one of the most important drug transporters in the intestine allowing oral availability of GABA-related and other drugs and prodrugs. Furthermore, a recent report shows involvement of this transporter family, namely the PAT2 subtype, in the autosomal dominant inherited disorder iminoglycinuria (12).Unfortunately, up to now the exact three-dimensional structure of PAT1, the transmembrane domain topology, and the substrate binding site are unknown. More structural information of PAT1 would allow a better understanding of the molecular mechanisms of its function and drug interaction, which is so far being investigated only in classic transport studies. Mutational analysis of putative extracellular regions is a suitable tool to get the first clue into transmembrane organization and relevant amino acid residues (6). This approach should also elucidate the spatial organization of the extracellular loops. The present study was performed to identify functionally important extracellular cysteine residues and their involvement in disulfide bridges. The relevance of disulfide bonds for membrane protein function is mainly based on the stabilization of a proper three-dimensional structure. The correct conformation in turn is essential for trafficking, surface expression, stability, and transport function. So far, intramolecular disulfide bonds have been identified for only very few SLCs, e.g. the serotonin transporter SERT and the dopamine transporter DAT (1315). Native disulfide bonds are probably required for transporter function of the Na+/glucose cotransporter SGLT1 (16, 17). For the type IIa sodium/phosphate cotransporter, it was shown that cleavage of disulfide bonds results in conformational changes that lead to internalization and subsequent lysosomal degradation of the transport protein (18). A similar stabilizing effect of an intramolecular disulfide bridge was also reported for the human ATP-binding cassette (ABC) transporter ABCG2 (19).Linkage via cysteine residues can also be necessary for transporter oligomerization. For the rat serotonin transporter SERT (20) and for the human ABC transporter ABCG2 (21), intermolecular disulfide bridges could be identified. For the hexose transporter GLUT1, an intramolecular disulfide bond promotes tetramerization of the transporter (22, 23). On the other hand, removal of cysteine residues can also lead to an impaired trafficking and mislocalization of the transporter protein without a disulfide bridge being involved (13, 24, 25). In those cases, the cysteine residues themselves are assumed to play an important role for the trafficking and targeting of the transporter to the cell surface. Similarly, for several transporters, cysteine residues located in a transmembrane domain play a key role in substrate recognition. Single cysteines have been found to be essential for substrate binding of the rat organic cation transporters rOCT1 and rOCT2 (26) and the multidrug and toxin extrusion transporter MATE1 (27). The relevance of conserved cysteines for the integrity of a membrane protein has therefore to be investigated very thoroughly. Several earlier studies reported loss of function in cysteine mutants without testing membrane localization.After assessing a negative influence of the reducing reagent DTT on hPAT1 function, we performed systematic mutagenesis in this study. The three putative extracellular cysteine residues Cys-180, Cys-329, and Cys-473 were individually exchanged to either alanine or serine residues. The resulting mutants were analyzed for substrate binding and transport in human retinal pigment epithelial (HRPE) cells and electrogenic transport in Xenopus laevis oocytes. Biochemical approaches provided direct evidence for an essential disulfide bond between Cys-180 and Cys-329. A triple mutant was constructed and examined to exclude other juxtamembrane cysteine residues as potential partners for disulfide bridges. The data suggest that this disulfide bridge is involved in forming or stabilizing the putative substrate-binding pocket. In addition, our results strongly support the eleven transmembrane domain topology model of hPAT1. This is consistent with our recently published data on glycosylation of hPAT1 (28).  相似文献   

8.
<正> DL—1—(2,6—二甲基苯氧基)—乙—氨基丙烷盐酸盐,商品名为慢心律(Mcxiletine)是七十年代进行临床研究的一个抗心律失常药物。目前国内外广泛用于临床,其特点是可以长期口服治疗和预防室性心律失常。根据国内外近十年的临床观察,若用量超过600—800mg/日,则有明显副作用如神经兴奋、噁心、复视、抽搐、振颤等。为了寻找更理想的抗心律失常药物,国内外已做过一些慢心律的类似物供药理筛选研究。本文是从前体药物的角度以慢心律自由碱DL—1—(2,6—甲基苯氧基)—2—氨基丙  相似文献   

9.
10.
11.
12.
Excitatory amino acid transporter (EAAT) glutamate transporters function not only as secondary active glutamate transporters but also as anion channels. Recently, a conserved aspartic acid (Asp112) within the intracellular loop near to the end of transmembrane domain 2 was proposed as a major determinant of substrate-dependent gating of the anion channel associated with the glial glutamate transporter EAAT1. We studied the corresponding mutation (D117A) in another EAAT isoform, EAAT4, using heterologous expression in mammalian cells, whole cell patch clamp, and noise analysis. In EAAT4, D117A modifies unitary conductances, relative anion permeabilities, as well as gating of associated anion channels. EAAT4 anion channel gating is characterized by two voltage-dependent gating processes with inverse voltage dependence. In wild type EAAT4, external l-glutamate modifies the voltage dependence as well as the minimum open probabilities of both gates, resulting in concentration-dependent changes of the number of open channels. Not only transport substrates but also anions affect wild type EAAT4 channel gating. External anions increase the open probability and slow down relaxation constants of one gating process that is activated by depolarization. D117A abolishes the anion and glutamate dependence of EAAT4 anion currents and shifts the voltage dependence of EAAT4 anion channel activation by more than 200 mV to more positive potentials. D117A is the first reported mutation that changes the unitary conductance of an EAAT anion channel. The finding that mutating a pore-forming residue modifies gating illustrates the close linkage between pore conformation and voltage- and substrate-dependent gating in EAAT4 anion channels.  相似文献   

13.
Germline mutations in DNA repair genes are linked to tumor progression. Furthermore, failure in either activating a DNA damage checkpoint or repairing programmed meiotic double-strand breaks (DSBs) can impair chromosome segregation. Therefore, understanding the molecular basis for DNA damage response (DDR) and DSB repair (DSBR) within the germline is highly important. Here we define ZTF-8, a previously uncharacterized protein conserved from worms to humans, as a novel factor involved in the repair of both mitotic and meiotic DSBs as well as in meiotic DNA damage checkpoint activation in the C. elegans germline. ztf-8 mutants exhibit specific sensitivity to γ-irradiation and hydroxyurea, mitotic nuclear arrest at S-phase accompanied by activation of the ATL-1 and CHK-1 DNA damage checkpoint kinases, as well as accumulation of both mitotic and meiotic recombination intermediates, indicating that ZTF-8 functions in DSBR. However, impaired meiotic DSBR progression partially fails to trigger the CEP-1/p53-dependent DNA damage checkpoint in late pachytene, also supporting a role for ZTF-8 in meiotic DDR. ZTF-8 partially co-localizes with the 9-1-1 DDR complex and interacts with MRT-2/Rad1, a component of this complex. The human RHINO protein rescues the phenotypes observed in ztf-8 mutants, suggesting functional conservation across species. We propose that ZTF-8 is involved in promoting repair at stalled replication forks and meiotic DSBs by transducing DNA damage checkpoint signaling via the 9-1-1 pathway. Our findings define a conserved function for ZTF-8/RHINO in promoting genomic stability in the germline.  相似文献   

14.
Plant architecture is influenced by the polar, cell-to-cell transport of auxin that is primarily provided and regulated by plasma membrane efflux catalysts of the PIN-FORMED and B family of ABC transporter (ABCB) classes. The latter were shown to require the functionality of the FK506 binding protein42 TWISTED DWARF1 (TWD1), although underlying mechanisms are unclear. By genetic manipulation of TWD1 expression, we show here that TWD1 affects shootward root auxin reflux and, thus, downstream developmental traits, such as epidermal twisting and gravitropism of the root. Using immunological assays, we demonstrate a predominant lateral, mainly outward-facing, plasma membrane location for TWD1 in the root epidermis characterized by the lateral marker ABC transporter G36/PLEIOTROPIC DRUG-RESISTANCE8/PENETRATION3. At these epidermal plasma membrane domains, TWD1 colocalizes with nonpolar ABCB1. In planta bioluminescence resonance energy transfer analysis was used to verify specific ABC transporter B1 (ABCB1)–TWD1 interaction. Our data support a model in which TWD1 promotes lateral ABCB-mediated auxin efflux via protein–protein interaction at the plasma membrane, minimizing reflux from the root apoplast into the cytoplasm.  相似文献   

15.
Virus-like particles (VLPs) have not been observed in Caenorhabditis germ cells, although nematode genomes contain low numbers of retrotransposon and retroviral sequences. We used electron microscopy to search for VLPs in various wild strains of Caenorhabditis, and observed very rare candidate VLPs in some strains, including the standard laboratory strain of C. elegans, N2. We identified the N2 VLPs as capsids produced by Cer1, a retrotransposon in the Gypsy/Ty3 family of retroviruses/retrotransposons. Cer1 expression is age and temperature dependent, with abundant expression at 15°C and no detectable expression at 25°C, explaining how VLPs escaped detection in previous studies. Similar age and temperature-dependent expression of Cer1 retrotransposons was observed for several other wild strains, indicating that these properties are common, if not integral, features of this retroelement. Retrotransposons, in contrast to DNA transposons, have a cytoplasmic stage in replication, and those that infect non-dividing cells must pass their genomic material through nuclear pores. In most C. elegans germ cells, nuclear pores are largely covered by germline-specific organelles called P granules. Our results suggest that Cer1 capsids target meiotic germ cells exiting pachytene, when free nuclear pores are added to the nuclear envelope and existing P granules begin to be removed. In pachytene germ cells, Cer1 capsids concentrate away from nuclei on a subset of microtubules that are exceptionally resistant to microtubule inhibitors; the capsids can aggregate these stable microtubules in older adults, which exhibit a temperature-dependent decrease in egg viability. When germ cells exit pachytene, the stable microtubules disappear and capsids redistribute close to nuclei that have P granule-free nuclear pores. This redistribution is microtubule dependent, suggesting that capsids that are released from stable microtubules transfer onto new, dynamic microtubules to track toward nuclei. These studies introduce C. elegans as a model to study the interplay between retroelements and germ cell biology.  相似文献   

16.
Mutations in PHF6 are the cause of B?rjeson-Forssman-Lehman syndrome (BFLS), an X-linked intellectual disability (XLID) disorder, and both T-cell acute lymphoblastic leukemia (T-ALL) and acute myeloid leukemia (AML). The PHF6 gene encodes a protein with two plant homeodomain (PHD)-like zinc finger domains. As many PHD-like domains function to target chromatin remodelers to post-translationally modified histones, this suggests a role for PHF6 in chromatin regulation. However, PHD domains are usually found in association with a catalytic domain, a feature that is lacking in PHF6. This distinct domain structure and the minimal information on its cellular function prompted us to perform a proteomic screen to identify PHF6 binding partners. We expressed recombinant Flag-tagged PHF6 in HEK 293T cells for coimmunoprecipitation, and analyzed the purified products by mass spectrometry. We identified proteins involved in ribosome biogenesis, RNA splicing, and chromatin regulation, consistent with PHF6 localization to both the nucleoplasm and nucleolus. Notably, PHF6 copurified with multiple constituents of the nucleosome remodeling and deacetylation (NuRD) complex, including CHD4, HDAC1, and RBBP4. We demonstrate that this PHF6-NuRD complex is not present in the nucleolus but is restricted to the nucleoplasm. The association with NuRD represents the first known interaction for PHF6 and implicates it in chromatin regulation.  相似文献   

17.
The glutamate transporter excitatory amino acid carrier 1 (EAAC1) catalyzes the co-transport of three Na+ ions, one H+ ion, and one glutamate molecule into the cell, in exchange for one K+ ion. Na+ binding to the glutamate-free form of the transporter generates a high affinity binding site for glutamate and is thus required for transport. Moreover, sodium binding to the transporters induces a basal anion conductance, which is further activated by glutamate. Here, we used the [Na+] dependence of this conductance as a read-out of Na+ binding to the substrate-free transporter to study the impact of a highly conserved amino acid residue, Thr101, in transmembrane domain 3. The apparent affinity of substrate-free EAAC1 for Na+ was dramatically decreased by the T101A but not by the T101S mutation. Interestingly, in further contrast to EAAC1WT, in the T101A mutant this [Na+] dependence was biphasic. This behavior can be explained by assuming that the binding of two Na+ ions prior to glutamate binding is required to generate a high affinity substrate binding site. In contrast to the dramatic effect of the T101A mutation on Na+ binding, other properties of the transporter, such as its ability to transport glutamate, were impaired but not eliminated. Our results are consistent with the existence of a cation binding site deeply buried in the membrane and involving interactions with the side chain oxygens of Thr101 and Asp367. A theoretical valence screening approach confirms that the predicted site of cation interaction has the potential to be a novel, so far undetected sodium binding site.  相似文献   

18.
Li ZC  Bush DR 《Plant physiology》1991,96(4):1338-1344
Proton-coupled aliphatic, neutral amino acid transport was investigated in plasma membrane vesicles isolated from sugar beet (Beta vulgaris L., cv Great Western) leaves. Two neutral amino acid symport systems were resolved based on inter-amino acid transport competition and on large variations in the specific activity of each porter in different species. Competitive inhibition was observed for transport competition between alanine, methionine, glutamine, and leucine (the alanine group) and between isoleucine, valine, and threonine (the isoleucine group). The apparent Km and Ki values were similar for transport competition among amino acids within the alanine group. In contrast, the kinetics of transport competition between these two groups of amino acids did not fit a simple competitive model. Furthermore, members of the isoleucine group were weak transport antagonists of the alanine group. These results are consistent with two independent neutral amino acid porters. In support of that conclusion, the ratio of the specific activity of alanine transport versus isoleucine transport varied from two- to 13-fold in plasma membrane vesicles isolated from different plant species. This ratio would be expected to remain relatively stable if these amino acids were moving through a single transport system and, indeed, the ratio of alanine to glutamine transport varied less than twofold. Analysis of the predicted structure of the aliphatic, neutral amino acids in solution shows that isoleucine, valine, and threonine contain a branched methyl or hydroxyl group at the β-carbon position that places a dense electron cloud close to the α-amino group. This does not occur for the unbranched amino acids or those that branch further away, e.g. leucine. We hypothesize that this structural feature of isoleucine, valine, and threonine results in unfavorable steric interactions with the alanine transport system that limits their flux through this porter. Hydrophobicity and hydrated volumes did not account for the observed differences in transport specificity.  相似文献   

19.
Target sequences for the C. elegans transposable element Tc1.   总被引:14,自引:3,他引:14       下载免费PDF全文
The target sequences for two independent insertions of the transposable element Tc1 from Caenorhabditis elegans show homology. Because both insertions are at palindromic TA/AT sequences, the exact boundaries of Tc1 cannot be distinguished; Tc1 could be 1610 bp and flanked by a 2-bp duplication of the target site or it could be 1612 bp and without target site duplication. The latter possibility implies a novel manner for insertion of a transposable element.  相似文献   

20.
Secondary transporters in the excitatory amino acid transporter family terminate glutamatergic synaptic transmission by catalyzing Na+-dependent removal of glutamate from the synaptic cleft. Recent structural studies of the aspartate-specific archaeal homolog, GltPh, suggest that transport is achieved by a rigid body, piston-like movement of the transport domain, which houses the substrate-binding site, between the extracellular and cytoplasmic sides of the membrane. This transport domain is connected to an immobile scaffold by three loops, one of which, the 3–4 loop (3L4), undergoes substrate-sensitive conformational change. Proteolytic cleavage of the 3L4 was found to abolish transport activity indicating an essential function for this loop in the transport mechanism. Here, we demonstrate that despite the presence of fully cleaved 3L4, GltPh is still able to sample conformations relevant for transport. Optimized reconstitution conditions reveal that fully cleaved GltPh retains some transport activity. Analysis of the kinetics and temperature dependence of transport accompanied by direct measurements of substrate binding reveal that this decreased transport activity is not due to alteration of the substrate binding characteristics but is caused by the significantly reduced turnover rate. By measuring solute counterflow activity and cross-link formation rates, we demonstrate that cleaving 3L4 severely and specifically compromises one or more steps contributing to the movement of the substrate-loaded transport domain between the outward- and inward-facing conformational states, sparing the equivalent step(s) during the movement of the empty transport domain. These results reveal a hitherto unknown role for the 3L4 in modulating an essential step in the transport process.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号